Acute kidney injury in hospitalized patients with liver

0 downloads 0 Views 3MB Size Report
cirrhosis: real life use of the new proposed diagnostic criteria .... in 1970, Epsteins' photographs of renal angiography pre- ..... Kidney Int 2009; 76: 422-427. 5.
Quarterly of the Polish Association for the Study of the Liver

Suppl 1/2017 ISSN | 2392-1099

Editors-in-chief Krzysztof Tomasiewicz, MD, PhD, DSc

Prof. Robert Flisiak, MD, PhD, DSc

Chair and Department of Infectious Diseases Medical University of Lublin e-mail: [email protected]

Department of Infectious Diseases and Hepatology Medical University of Bialystok e-mail: [email protected]

Editorial office TERMEDIA Publishing House Kleeberga 2, 61-615 Poznań, Poland phone/fax: +48 61 822 77 81 e-mail: [email protected]

Editorial Board Alessio Aghemo, MD, PhD (Milan) Prof. Tarik Asselah (Paris) Jordan Feld, MD, MPH (Toronto) Prof. Andrzej Gabriel, MD, PhD, DSc (Zabrze) Prof. Andrzej Habior, MD, PhD, DSc (Warsaw) Prof. Waldemar Halota, MD, PhD, DSc (Bydgoszcz) Prof. Bela Hunyady, MD, PhD, DSc (Kaposvár) Maciej Jabłkowski, MD, PhD, DSc (Lodz) Prof. Peter Jarcuska (Košice) Jerzy Jaroszewicz, MD, PhD, DSc (Bytom) Maciej Jabłkowski, MD, PhD, DSc (Lodz) Michal Kukla, MD, PhD, DSc (Katowice) Prof. Piotr Małkowski, MD, PhD, DSc (Warsaw) Michael P. Manns, MD (Hannover) Włodzimierz Mazur, MD, PhD, DSc (Chorzow) Prof. Jean-Michel Pawlotsky, MD, PhD (Creteil) Prof. Joanna Pawłowska, MD, PhD, DSc (Warsaw) Prof. Małgorzata Pawłowska, MD, PhD, DSc (Bydgoszcz) Anna Piekarska, MD, PhD, DSc (Lodz) Prof. Krzysztof Simon, MD, PhD, DSc (Wroclaw) Bożena Walewska-Zielecka, MD, PhD, DSc (Warsaw) Prof. Marek Woynarowski, MD, PhD, DSc (Warsaw)

Publisher TERMEDIA Publishing House e-mail: [email protected] http://www.termedia.pl Poznan office Kleeberga 2, 61-615 Poznań, Poland phone/fax: +48 61 822 77 81 Warsaw office phone/fax: +48 22 827 75 14 e-mail: [email protected]

Quarterly of the Polish Association for the Study of the Liver

President of the Management Board Janusz Michalak Scientific Director Maciej Banach Production Editor Marzena Demska; e-mail: [email protected] Marketing and Advertising Department Mikołaj Lasocki; e-mail: [email protected]

Formerly Experimental & Clinical Hepatology, ISSN 1734-3038

The journal is originally published in the print version. Copyright: © 2017 Clinical and Experimental Hepatology. This is an Open Access journal, all articles are distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/ licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.

Suppl 1/2017: s1–s36

Contents Original paper

Acute kidney injury in hospitalized patients with liver cirrhosis: real life use of the new proposed diagnostic criteria.................................................................s1 Lubomir Skladany, Juraj Svac, Lukas Liptak, Daniela Jancekova, Janka Vnencakova, Svetlana Adamcova-Selcanova Review paper

Acute-over-chronic liver failure (ACLF) – immunity, infection, inflammation ...........................s7 Lubomir Skladany, Svetlana Adamcova-Selcanova, Daniela Jancekova

9th Symposium on Portal Hypertension The liver and infections .................................................................................................................. s17 Programme ........................................................................................................................................ s22 Oral section ........................................................................................................................................ s24 Poster section ..................................................................................................................................... s34

Clin Exp HEPATOL Suppl 1/2017; s1–s6

Original paper

Acute kidney injury in hospitalized patients with liver cirrhosis: real life use of the new proposed diagnostic criteria Lubomir Skladany1, Juraj Svac1, Lukas Liptak1, Daniela Jancekova1, Janka Vnencakova1,2, Svetlana Adamcova-Selcanova1 Department of Internal Medicine II, Slovak Medical University, F.D. Roosevelt University Hospital, Banská Bystrica, Slovakia Faculty of Healthcare, Slovak Medical University, F.D. Roosevelt University Hospital, Banská Bystrica, Slovakia

1 2

Abstract Introduction: Acute kidney injury (AKI) in liver cirrhosis (ACLD) is frequent and associated with increased mortality; it could appear far earlier than full-blown hepatorenal syndrome (HRS) evolves. It was of much interest to the hepatology, therefore, when nephrologists begun to refine diagnostic criteria of what is currently known as AKI. Attempts to adopt it for patients with ACLD have led to widespread discussion; most controversies were related to leaving out of (already lowered) threshold creatininaemia (SCr) of 133 µmol/l for the definition of HRS type I. The ensuing new proposed diagnostic criteria for the AKI in ACLD have been in the process of external validation; therefore, studies from the real life clinical practice are of interest. The aim of this study was to investigate their applicability and prognostic validity in the cohort of hospitalized patients with decompensated ACLD (dACLD). Material and methods: Retrospective study of consecutive admissions to tertiary referral liver unit. Inclusion criteria: hospital admission with dACLD; exclusion criteria: malignancy other than HCC, lack of data for analysis. Study interval: January 1st, 2013 – September 30th, 2013. Definitions: AKI – the new proposed criteria according to International Club of Ascites (ICA-AKI); chronic kidney disease (CKD) was defined according to KDIGO criteria. Results: During study interval of 9 months, 326 patients with dACLD were admitted; females – 38%, mean age – 53 years (y), MELD – 16.5, ascites (CiA) – 47%. Etiology of dACLD: alcoholic liver disease (ALD) – 55%. AKI was diagnosed in 26% of dACLD admissions; AKI – stage 1 in 14%, AKI – stage 2 in 6%, and AKI – stage 3 in 6%, respectively. CKD, present in 26%, was the risk-factor for AKI. In-hospital mortality was 21% in AKI vs. 1% in non-AKI group, respectively (p < 0.0001); in AKI-1a (SCr < 133 µmol/l) 2.6%, AKI-1b 21.2% (p = 0.01). Complications were recorded in 39% of patients with AKI vs. 7% in non-AKI group, respectively (p < 0.0001); AKI-1a – 21%, AKI-1b – 33% (p = 0.24). Conclusions: This study has shown that new proposed diagnostic criteria for AKI in patients with dACLD are applicable in daily clinical practice outside the context of clinical trials. The main significance of adopting new diagnostic criteria dwells in refined prognostic stratification of patients with dACLD, earlier introduction of volumexpansion with albumin, earlier introduction of terlipressin if albumin alone does not lead to improvement of AKI, and more personalized approach. In this study AKI-1a with SCr of < 133 µmol/l was associated with complications, but not with mortality. Key words: liver cirrhosis, advanced chronic liver disease, acute kidney injury, diagnostic criteria.

Address for correspondence Lubomir Skladany, MD, PhD, Head, Department of Internal Medicine II, Slovak Medical University, F.D. Roosevelt University Hospital, Nam. L. Svobodu 1, 97401 Banská Bystrica, e-mail: [email protected]

Clinical and Experimental Hepatology Suppl 1/2017

s1

Lubomir Skladany, Juraj Svac, Lukas Liptak, Daniela Jancekova, Janka Vnencakova, Svetlana Adamcova-Selcanova

Introduction In 2012, the estimated worldwide toll of acute kidney injury (AKI) would be 3 million deaths [1-3]. Every other patient admitted to intensive care unit (ICU) develop AKI, and 27% die before hospital discharge (four times that of patients without AKI) [3-6]. In liver cirrhosis, nowadays in clinical practice called advanced chronic liver disease (ACLD), AKI is frequent and deadly [4]. On admission, it is present in 19-32% of patients with dACLD (community-acquired AKI), and in 52% if acute chronic liver failure (ACLF) is present; the mortality of AKI stages 1, 2 and 3 is around 15%, 30%, and 60%, respectively [7-9]. The diagnostic criteria and management of AKI in ACLD are being under epochal transformation [10-12]. Although the term ‘hepatorenal syndrome’ (HRS) was already used in the first-third of 20th century, socalled ‘traditional diagnostic criteria’ were established only in 1996 and updated in 2007. In between, in 1959, Papper has described extreme renal vasoconstriction; in 1963, Flint published first description of the syndrome; in 1970, Epsteins’ photographs of renal angiography preand post-mortem flew around the world, and in 1975, Rodes divided kidney injury into three types [13-20]. Acute renal failure (ARF), the term predeceasing AKI, was defined as the increase of serum creatinine (sCr) level of ≥ 50% from baseline and to ≥ 133 μmol/l [13, 19]. These diagnostic criteria have been a  strong predictor of in-hospital mortality, and have had a high specificity, but suffered from low sensitivity leading to late diagnosis and limited response to treatment in some patients [21-24]. Meanwhile, researchers as well as clinicians have recognized weaknesses of all three cornerstones of traditional diagnostic criteria for ARF in ACLD: threshold creatininaemia, timeframe of change in sCr, and sCr itself – as a biomarker of renal function [10, 11]. 1. The use of diagnostic threshold for ARF/AKI in ACLD (sCr ≥ 133 μmol/l) has been supported by strong evidence that it predicts progression of AKI and prognosis of patients [7, 25]. However, the threshold had serious limitations: i) AKI was diagnosed in relatively late stage, hence could be less responsive to the therapy [8]; ii) it does not take into account changes over days to weeks to discern AKI from chronic kidney disease (CKD); iii) AKI with sCr < 133 μmol/l decreased survival in the cases: a) precipitated by infection (i-AKI), b) progressing, and c) community-acquired AKI [26-30]. 2. Timeframe used for diagnosing ARF/AKI in ACLD, except for HRS 1, often does not allow for unequivocal distinction between AKI and CKD. This point relates closely to the definition of baseline sCr, which

s2

if strict (i.e., 7 days, 14 days etc.), is destined to be under used in real-life clinical practice, where it is often unavailable. 3. Last but not least, there is a problem of sCr as a biomarker of renal function in general and in ACLD in particular: it is influenced by age, gender, sarcopenia, increased tubular secretion of creatinine, increased volume of distribution, interference of bilirubin with assays, etc. [31-33]. Overall, in ACLD, sCr overestimates projected glomerular filtration rate (eGFR) or kidney function [10, 11]. These drawbacks have fueled endeavors to redefine ARF in general population. Two separate bodies developed two consensus definitions of AKI (the term ARF was officially abandoned): the Acute Dialysis Quality Initiative group issued so-called RIFLE criteria (Risk, Injury, Failure, End-stage renal disease), and the Acute Kidney Injury Network (AKIN), so-called AKIN criteria of AKI [34-36]. Subsequently, panel of experts has suggested combining these two sets of criteria into unifying KDIGO criteria for AKI (the Kidney Disease Improving Global Outcome) [35]. These criteria were then tested in patients with ACLD and discussed on the Venice Convention of International Club of Ascites (ICA) in 2012; the discussion continued until April 2015, when experts agreed on the final proposal of a new approach to the diagnosis and therapy of AKI in cirrhosis (new proposed criteria) [7, 25, 27-29, 37-40]. The main difference from traditional criteria was the abandonment of fixed thresholds of sCr ≥ 133 μmol/l and > 221 μmol/l for AKI and HRS-1, respectively (Table 1); this is important, since the higher sCr, the lower the probability of response to terlipressin with albumin [41, 42]. Proof of concept for the new proposed criteria has been provided by their ability to predict mortality in various settings: i) in hospitalized patients with liver cirrhosis including those on ICU [7, 25, 27, 29, 38-40, 43-45]; ii) in cases of AKI with progression [7, 25, 39]; iii) in AKI grade 2 and 3 [7, 25, 39]; iv) in AKI with the highest sCr < 133 μmol/l (AKI-1a) [29, 30]; v) in outpatients with resolution of AKI [27]. The main lesson learnt from the application of the new proposed criteria was that even small increase in sCr should be identified for potentially early intervention. Only recommendation for treatment with vasoconstrictors of AKI-1a without change after volumexpansion with albumin requires more investigation [10, 11, 26]. The new proposed criteria do not eliminate the possibility of parenchymal renal disease in HRS; however, the potential role of urinary biomarkers in this setting is still unclear [7, 39, 46]. Response to therapy is classified as full, partial, and none with prognostic implications [47].

Clinical and Experimental Hepatology Suppl 1/2017

Acute kidney injury in hospitalized patients with liver cirrhosis: real life use of the new proposed diagnostic criteria

Table 1. International Club of Ascites (ICA-AKI) new definitions for the diagnosis and management of acute kidney injury (AKI) in patients with cirrhosis [10, 11] Subject

Definition

Baseline sCr

A value of sCr obtained in the previous 3 months, when available, can be used as baseline sCr. In patients with more than one value within the previous 3 months, the value closest to the admission time to the hospital should be used. In patients without a previous sCr value, the sCr on admission should be used as baseline.

Definition of AKI

«2WL[NJ\NRW\,[sVPMUstVXUU`R]QRW!QX^[\$X[ «*YN[LNW]JPNRWL[NJ\N\,[sO[XVKJ\NURWN`QRLQR\TWX`WX[Y[N\^VNM]XQJ_NXLL^[[NM`R]QRW]QNY[RX[ MJb\

Staging of AKI

«Stage 1#RWL[NJ\NRW\,[sVPMUtVXUUX[JWRWL[NJ\NRW\,[sOXUM]XOXUMO[XVKJ\NURWN «Stage 2: increase in sCr > 2-fold to 3-fold from baseline «Stage 3#RWL[NJ\NXO\,['OXUMO[XVKJ\NURWNX[\,[sVPMUtVXUU`R]QJWJL^]NRWL[NJ\NsVPMU (26.5 µmol/l) or initiation of renal replacement therapy

Progression of AKI Response to treatment

Progression Progression of AKI to a higher stage and/or need for RRT No response No regression of AKI

Partial response Regression of AKI stage with a reduction XO\,[]XsVPMUtVXUUJKX_N the baseline value

The aim of the study was to investigate the applicability and prognostic relevance of the new proposed ICA-AKI diagnostic criteria in real-life clinical setting.

Material and methods Single center study at the division of Hepatology, Gastroenterology, and Liver Transplantation (HEGITO) of the Department of Internal Medicine in the Regional Teaching Hospital. Retrospective analysis of database ‘HEGITO 7’, which collects data of patients hospitalized with ACLD. Inclusion criteria: hospital admission for (dACLD); exclusion criteria: malignancy other than HCC, lack of data for analysis. Study interval: January 1st, 2013 – September 30th, 2013. As definitions for AKI and CKD, ICA-AKI and KDIGO criteria were used, respectively (Table 1) [10, 11, 35]. As a baseline sCr for diagnosis of AKI, values on admission or < 3 months prior to admission have been used. These were related to further three of sCr readings during hospitalization: minimal, maximal, and at discharge. Other recorded variables: age, gender, etiology of dACLD, drugs used before admission (specifically furosemide, spironolactone, angiotensin converting enzyme inhibitors and sartans), model for end stage liver disease (MELD), in-hospital mortality, complications of ACLD: ascites, as defined by [48], infections [49], variceal bleeding, hepatic encephalopathy [50], and length of hospital stay (LOS). Management of AKI followed traditional criteria and EASL guidelines of 2010 [48]. Notably, patients hospitalized for dACLD with AKI on admission have received volumexpansion with crystalloids and albumin 20%; those with HRS-1 with albumin 20% plus terlipressin after 48 hours in albumin non-responders.

Clinical and Experimental Hepatology Suppl 1/2017

Regression Regression of AKI to a lower stage Full response Return of sCr to a value within 0.3 mg/dl (26.5 µmol/l) of the baseline value

The administration of terlipressin was intravenous boluses q6h, in doses according to body weight: 1 mg or 2 mg in patients with < or > 70 kg, respectively [4, 5, 17]. Primary endpoints: prevalence of AKI according to new proposed criteria, in-hospital mortality related to AKI stages. Secondary endpoints: association of AKI with complications and LOS; differences between AKI-1a and -1b. Statistical analysis: descriptive, parametric variables were summarized by mean and standard deviation, and compared by Student‘s t-test, categorical variables were compared using c2 test. Two-sided null hypotheses of no difference were rejected if p-values were less than 0.05. Cases with missing data were allowed zero value or discarded from analysis.

Results From 972 hospitalizations during study interval of 9 months, 326 were for dACLD (33%) (Table 2); females – 123 (37.7%); mean age – 53 years (19-85); MELD – 16.5 (6.6-46.7); ascites – 155 (47.5%); HCC – 21 (6.4%); laboratory values on admission: albumin – 31.8 ± 6.9 g/l; bilirubin – 86.6 µmol/l (4.7-670.6); sCr – 103.5 µmol/l (37-609); international normalized ratio (INR) – 1.51 ± 0.36; prothrombin time (Quick, PT) – 52 ± 14.5%. Etiology of dACLD: alcoholic liver disease (ALD) 179 (54.9%); primary biliary and sclerosing cholangitis (PBC + PSC) 26 (8%); hepatitis B and C viral infection (HBV + HCV) 24 (7.3%). AKI was diagnosed in 110 of 326 cases (33.7%); AKI-1 in 71 (21.7%), AKI-2 in 19 (5.8%), and AKI-3 in 20 cases (6.1%). MELD scores calculated on admission in AKI vs. non-AKI groups were 21.8 (8.2-46.7) vs. 14.6 (6.2-43.1) (p < 0.0001); MELD in AKI-1a vs. AKI-1b were 17.4 (8.4-36.8) vs. 23.1

s3

Lubomir Skladany, Juraj Svac, Lukas Liptak, Daniela Jancekova, Janka Vnencakova, Svetlana Adamcova-Selcanova

Table 2. Prevalence of acute kidney injury according to baseline demographic and clinical variables (n = 326) Variable

No AKI

AKI

Number of patients

242

84

Age (years)

53.9

54.4

n.s.

Gender female (%)

26.7

11

n.s.

ALD (%)

67

33

0.001

Viral hepatitis B + C (%)

88

13

Other etiologies (%)

84

16

MELD score (points)

14.6

21.8

p

0.5 0.4 p = 0.01

0.0004

(10.4-40.2) (p = 0.0076). AKI was more prevalent in ALD as compared to both viral, and other etiologies of ACLD (33%, 12%, 16%, respectively; p = 0.001). In-hospital mortality (n = 216) (Fig. 1): there were 20 deaths in AKI (21%), and 2 (1%) in non-AKI groups, respectively (p < 0.0001); mortality in AKI subgroups: AKI-1 – 8/61 (13.1%) (AKI-1a – 1/38 (2.6%), AKI-1b – 7/33 (21.2%), p = 0.01), AKI-2 – 4/19 (21.0%) (p vs. AKI-1b = 0.99), AKI-3 – 11/20 (55%) (p vs. AKI-2 = 0.03, p vs. non-AKI = 0.00001). Complications (Fig. 2): frequency in AKI vs. non-AKI group was 43/110 (39%) vs. 16/216 (7.4%) (p < 0.0001); in AKI-1 – 19/71 (26.7%) (AKI-1a 8/38 (21.1%), AKI 1b 11/33 (33%) (p = 0.24), in AKI-2 – 12/19 (63%) (p vs. AKI-1b = 0.04), in AKI-3 – 11/20 (60%) (p vs. non-AKI = 0.000001, p vs. AKI-2 = 0.84). Mean LOS in whole cohort was 9.5 days (1-45) (Fig. 3). LOS in non-AKI vs. AKI were 6.8 days (1-34) vs. 14.7 days (0-45) (p < 0.0001); in AKI-1 LOS was 12.3 days (AKI-1a – 12.4 days, AKI-1b – 12.1 days) (p = 0.88), in AKI-2 – 17.6 days (p vs. AKI-1b = 0.003), AKI-3 20.9 days (p vs. non-AKI = 0.000001, p vs. AKI-2 = 0.0005). CKD on admission: overall – 86/326 (26.4%), grade 2 (G2) – 58 (17.8%), grade 3 (G3) – 21 (6.4%), grade 4 (G4) – 6 (1.8%), grade 5 p = 0.000001

0.2

p = 0.24

p = 0.04

0.3

AKI-1b

AKI-2

AKI-3

Discussion As in several other papers on the topic, results of current study support the case for implementing the new proposed ICA-AKI criteria into daily clinical practice. Thanks to these criteria, even small changes in sCr – otherwise often left unrecognized due mainly to low baseline sCr in patients with ACLD – are being classified and, more importantly, treated as AKI [8, 25, 27-29, 37, 39, 40]. One-in-four prevalence of AKI in our patients hospitalized with dACLD compares well with 19-47% from other studies, as does its 14% – 6% – 6% distribution of AKI grades 1, 2, and 3. Lower age (51 years) and more ALD etiology of ACLD in our study are beyond the scope of discussion, since between-studies comparison of mortality and complications were not intended. It is apparent from the results, that the probability of AKI is etiology-of-ACLD – sensitive, with significantly higher prevalence in ALD than other etiologies. The most important finding has been the association of AKI ≥ 1b p

15

p = 0.84

AKI-1a

(G5) 1 (0.3%). In CKD ≤ G2 as compared to CKD > 2, the frequency of AKI was 23.8% (71/298) vs. 46.4% (13/28) (p = 0.009).

NS

0.4

No

Fig 1. Mortality according to acute kidney injury (AKI) grades (n = 326)

20

0.5

NS

0.1

25

0.6

p = 0.03

0.3

0

AKI – acute kidney injury, MELD – model for end stage liver disease, ALD – alcoholic liver disease

0.7

p = 0.00001

0.6

0.00025632

No vs. A1

0.003350856

No vs. B1

0.000477596

No vs. 2

8.53601E-05

No vs. 3

0.87886911

A1 vs. B1

0.079071804

B1 vs. 2

0.000477596

2 vs. 3

10

0.2 5

0.1 0

No

AKI-1a

AKI-1b

AKI-2

AKI-3

Fig 2. Complications according to acute kidney injury (AKI) grades (n = 326)

s4

0

No

AKI-1a

AKI-1b

AKI-2

AKI-3

Fig 3. Length of hospital stay (days) (n = 326)

Clinical and Experimental Hepatology Suppl 1/2017

Acute kidney injury in hospitalized patients with liver cirrhosis: real life use of the new proposed diagnostic criteria

with mortality (Fig. 1); notably, more than 50% of patients with AKI-3 has died. On the other hand, similar mortality of patients with AKI-1a as compared to patients without AKI could be seen as the argument against the abandonment of threshold sCr of 133 μmol/l (1.5 mg/ dl) [12, 51]. However, significantly higher complications (Fig. 2) in AKI-1a as compared to non-AKI group, allows for an alternative explanation: recognizing the earliest AKI stage leads to more sensitive approach to all the other complications of ACLD, their earlier recognition, more intense therapy, and reduction in mortality; the association of AKI-1a with complications is real and has been also supported by LOS, which was almost twice as high as in patients without AKI (Fig. 3). Of interest was 26% prevalence of CKD at admission; AKI was present in half the patients with CKD ≤ 2, as compared to CKD grade > 2; CKD should therefore be scrutinized as the possible risk factor for AKI. This study suffers several limitations. Bias introduced by its retrospective design was somewhat mitigated by the fact that in-hospital data has been collected over the span of the study in standardized and supervised manner (JV). The design has not allowed for distinguishing nosocomial AKI from community-acquired. Number of patients, although limited, has not precluded main conclusions concerning mortality, but could have influenced secondary endpoints, particularly differences between AKI-1a and 1b. Impossibility to retrieve detailed information on management, specifically the exact type and dose of volumexpanders, and the threshold for addition of terlipressin; this precluded the retrospective diagnosis of HRS, which is one of the major drawbacks of the study. However, according to the recent analysis of 300 patients with cirrhosis and AKI from Italy and Spain, the distribution of HRS among AKI stages 1-3 has been shown to be even; the main results of our study would stay unchanged [52]. The aim of this study was to investigate in real-life clinical practice the feasibility and relevance of the new proposed diagnostic criteria for AKI in ACLD shortly after their announcement in 2011 (the Berlin 2011 ICA-EASL Joint Meeting [Renal Dysfunction in Cirrhosis in the era of AKI: towards a new definition]) [26]. The results lend support to the lowering of threshold for the diagnosis of AKI. Even in its earliest stages, AKI is associated with worse outcome. It will be of interest to see the general impact of the new proposed diagnostic criteria on the outcome in the ACLD cohort, since they could lead to earlier interventions.

Conclusions The new proposed diagnostic criteria of ICA-AKI are applicable in ACLD patients from real-life clinical

Clinical and Experimental Hepatology Suppl 1/2017

practice. They were of immediate clinical relevance, since AKI of all stages was associated with complications and LOS. Mortality was associated only with AKI stages 1b and higher. As to the controversy surrounding AKI-1a, our results are difficult to interpret; for the abovementioned reasons, however, authors will retain AKI-1a as the stage most amenable to interventions with the probability of preventing fatal outcomes.

Disclosure Authors report no conflict of interest. References 1. Kellum JA, Lameire N; KDIGO AKI Guideline Work Group. Diagnosis, evaluation, and management of acute kidney injury: a KDIGO summary (Part 1). Crit Care 2013; 17: 204. 2. Kellum JA, Sileanu FE, Murugan R, et al. Classifying AKI by urine output versus serum creatinine level. J Am Soc Nephrol 2015; 26: 2231-2238. 3. Kellum JA, Murugan R. Effects of non-severe acute kidney injury on clinical outcomes in critically ill patients. Crit Care 2016; 20: 159. 4. Bouchard J, Soroko SB, Chertow GM, et al. Fluid accumulation, survival and recovery of kidney function in critically ill patients with acute kidney injury. Kidney Int 2009; 76: 422-427. 5. Hoste EAJ, Bagshaw SM, Bellomo R, et al. Epidemiology of acute kidney injury in critically ill patients: the multinational AKI-EPI study. Intensive Care Med 2015; 41: 1411-1423. 6. Uchino S, Kellum JA, Bellomo R, et al. Acute renal failure in critically ill patients: a multinational, multicenter study. JAMA 2005; 294: 813-818. 7. Fagundes C, Barreto R, Guevara M, et al. A modified acute kidney injury classification for diagnosis and risk stratification of impairment of kidney function in cirrhosis. J Hepatol 2013; 59: 474-481. 8. Garcia-Tsao G, Parikh CR, Viola A. Acute kidney injury in cirrhosis. Hepatology 2008; 48: 2064-2077. 9. Janicko M, Veseliny E, Senajova G, et al. Predictors of hepatorenal syndrome in alcoholic liver cirrhosis. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2015; 159: 661-665. 10. Angeli P, Gines P, Wong F, et al. Diagnosis and management of acute kidney injury in patients with cirrhosis: revised consensus recommendations of the International Club of Ascites. Gut 2015; 64: 531-537. 11. Angeli P, Gines P, Wong F, et al. Diagnosis and management of acute kidney injury in patients with cirrhosis: Revised consensus recommendations of the International Club of Ascites. J Hepatol 2015; 62: 968-974. 12. Arroyo V. Acute kidney injury (AKI) in cirrhosis: Should we change current definition and diagnostic criteria of renal failure in cirrhosis? J Hepatol 2013; 59: 415-417. 13. Arroyo V, Ginès P, Gerbes G, et al. Definition and diagnostic criteria of refractory ascites and hepatorenal syndrome in cirrhosis. Hepatology 1996; 23: 164-176. 14. Epstein M, Berk DP, Hollenberg NK, et al. Renal failure in the patient with cirrhosis. The role of active vasoconstriction. Am J Med 1970; 49: 175-185. 15. Flint A. Clinical report on hydro-peritoneum based on an analysis of forty-six cases. Am J Med Sci 1863; 45: 306-339.

s5

Lubomir Skladany, Juraj Svac, Lukas Liptak, Daniela Jancekova, Janka Vnencakova, Svetlana Adamcova-Selcanova

16. Hecker R, Sherlock S. Electrolyte and circulatory changes in terminal liver failure. Lancet 1956; 271: 1121-1125. 17. Papper S, Belsky JL, Bleifer KH. Renal failure in Laennec‘s cirrhosis of the liver. I. Description of clinical and laboratory features. Ann Intern Med 1959; 51: 759-773. 18. Rodés J, Bosch J, Arroyo V. Clinical types and drug therapy of renal impairment in cirrhosis. Postgrad Med J 1975; 51: 492-497. 19. Salerno F, Gerbes A, Ginés P, et al. Diagnosis, prevention and treatment of hepatorenal syndrome in cirrhosis. Gut 2007; 56: 1310-1318. 20. Schroeder ET, Shear L, Sancetta SM, et al. Renal failure in patients with cirrhosis of the liver. 3. Evaluation of intrarenal blood flow by para-aminohippurate extraction and response to angiotensin. Am J Med 1967; 43: 887-896. 21. Bataller R, Ginès P, Guevara M, et al. Hepatorenal syndrome. Semin Liver Dis 1997; 17: 233-247. 22. Francoz C, Glotz D, Moreau R, et al. The evaluation of renal function and disease in patients with cirrhosis. J Hepatol 2010; 52: 605-613. 23. Ginès P, Arroyo V, Vargas V, et al. Paracentesis with intravenous infusion of albumin as compared with peritoneovenous shunting in cirrhosis with refractory ascites. N Engl J Med 1991; 325: 829-835. 24. Sort P, Navasa M, Arroyo V, et al. Effect of intravenous albumin on renal impairment and mortality in patients with cirrhosis and spontaneous bacterial peritonitis. N Engl J Med 1999; 5: 403-409. 25. Piano S, Rosi S, Maresio G, et al. Evaluation of the Acute Kidney Injury Network criteria in hospitalized patients with cirrhosis and ascites. J Hepatol 2013; 59: 482-489. 26. Angeli P, Sanyal A, Moller S, et al. Current limits and future challenges in the management of renal dysfunction in patients with cirrhosis: report from the International Club of Ascites. Liver Int 2013; 33: 16-23. 27. Tsien CD, Rabie R, Wong F. Acute kidney injury in decompensated cirrhosis. Gut 2013; 62: 131-137. 28. Wong F, Nadim MK, Kellum JA, et al. Working Party proposal for a  revised classification system of renal dysfunction in patients with cirrhosis. Gut 2011; 60: 702-709. 29. Wong F, O’Leary JG, Reddy KR, et al. New consensus definition of acute kidney injury accurately predicts 30-day mortality in patients with cirrhosis and infection. Gastroenterology 2013; 145: 1280-1288. 30. Wong F, Murray P. Kidney damage biomarkers: novel tools for the diagnostic assessment of acute kidney injury in cirrhosis. Hepatology 2014; 60: 455-457. 31. Caregaro L, Menon F, Angeli P, et al. Limitations of serum creatinine level and creatinine clearance as filtration markers in cirrhosis. Arch Intern Med 1994; 154: 201-205. 32. Sherman DS, Fish DN, Teitelbaum I. Assessing renal function in cirrhotic patients: problems and pitfalls. Am J Kidney Dis 2003; 41: 269-278. 33. Spencer K. Analytical reviews in clinical biochemistry: the estimation of creatinine. Ann Clin Biochem 1986; 23 (Pt 1): 1-25. 34. Bellomo R, Ronco C, Kellum J, et al. Acute renal failure – definition, outcome measures, animal models, fluid therapy and information technology needs: The Second International Consensus Conference of the Acute Dialysis Quality Initiative (ADQI) Group. Crit Care 2004; 8: R204-R212. 35. KDIGO AKIWG. Kidney Disease: Improving Global Outcomes (KDIGO) Clinical Practice Guideline for Acute Kidney Injury. Kidney Inter Suppl 2012; 2: 1-138.

s6

36. Mehta RL, Kellum JA, Shah SV, et al. Acute Kidney Injury Network: report of an initiative to improve outcomes in acute kidney injury. Crit Care 2007; 11: R31. 37. Altamirano J, Fagundes C, Dominguez M, et al. Acute kidney injury is an early predictor of mortality for patients with alcoholic hepatitis. Clin Gastroenterol Hepatol 2012; 10: 65-71. 38. Angeli P, Rodríguez E, Piano S, et al. Acute kidney injury and acute-on-chronic liver failure classifications in prognosis assessment of patients with acute decompensation of cirrhosis. Gut 2015; 64: 1616-1622. 39. Belcher JM, Garcia-Tsao G, Sanyal AJ, et al. Association of AKI with mortality and complications in hospitalized patients with cirrhosis. Hepatology 2013; 57: 753-762. 40. de Carvalho JR, Villela-Nogueira CA, Luiz RR, et al. Acute kidney injury network criteria as a predictor of hospital mortality in cirrhotic patients with ascites. J Clin Gastroenterol 2012; 46: e21-e26. 41. Boyer TD, Sanyal AJ, Garcia-Tsao G, et al. Predictors of response to terlipressin plus albumin in hepatorenal syndrome (HRS) type 1: relationship of serum creatinine to hemodynamics. J Hepatol 2011; 55: 315-321. 42. Rodriguez E, Elia C, Solà E, et al. Terlipressin and albumin for type-1 hepatorenal syndrome associated with sepsis. J Hepatol 2014; 60: 955-961. 43. Joannidis M, Metnitz B, Bauer P, et al. Acute kidney injury in critically ill patients classified by AKIN versus RIFLE using the SAPS 3 database. Intensive Care Med 2009; 35: 1692-1702. 44. Pan HCh, Chien YS, Jenq ChCh, et al. Acute kidney injury classification for critically ill cirrhotic patients: A  comparison of the KDIGO, AKIN, and RIFLE classifications. Sci Rep 2016; 6: 23022. 45. Tu KH, Jenq CC, Tsai MH, et al. Outcome scoring systems for short-term prognosis in critically ill cirrhotic patients. Shock 2011; 36: 445-450. 46. Trawalé JM, Paradis V, Rautou PE, et al. The spectrum of renal lesions in patients with cirrhosis: a  clinicopathological study. Liver Int 2010; 30: 725-732. 47. Boyer TD, Sanyal AJ, Wong F, et al. Initial report of a  large, randomized, double blind, placebo-controlled, phase 3 trial of terlipressin plus albumin for the treatment of type 1 hepatorenal syndrome (HRS-1): the REVERSE study. Hepatology 2014; 60: 255A. 48. European Association for the Study of the Liver. EASL clinical practice guidelines on the management of ascites, spontaneous bacterial peritonitis, and hepatorenal syndrome in cirrhosis. J Hepatol 2010; 53: 397-417. 49. Fasolato S, Angeli P, Dallagnese L, et al. Renal failure and bacterial infections in patients with cirrhosis: epidemiology and clinical features. Hepatology 2007; 45: 223-229. 50. Vilstrup H, Amodio P, Bajaj J, et al. Hepatic encephalopathy in chronic liver disease: 2014 Practice Guideline by the American Association for the Study of Liver Diseases and the European Association for the Study of the Liver. Hepatology 2014; 60: 715-735. 51. Wong F, O‘Leary JG, Reddy KR, et al. A cut-off serum creatinine value of 1.5 mg/dl for AKI – to be or not to be. J Hepatol 2015; 62: 741-743. 52. Huelin P, Piano S, Solà E, et al. Validation of a Staging System for Acute Kidney Injury in Patients With Cirrhosis and Association With Acute-on-Chronic Liver Failure. Clin Gastroenterol Hepatol 2017; 15: 438-445.

Clinical and Experimental Hepatology Suppl 1/2017

Clin Exp HEPATOL Suppl 1/2017; s7–s15

Review paper

Acute-over-chronic liver failure (ACLF) – immunity, infection, inflammation Lubomir Skladany, Svetlana Adamcova-Selcanova, Daniela Jancekova Department of Internal Medicine II, Slovak Medical University, F.D. Roosevelt University Hospital, Banská Bystrica, Slovakia

Abstract The syndrome of acute-over-chronic liver failure (ACLF) was described in the landmark CANONIC study as the process resulting from the acute insult to the pre-existing chronic liver disease (ACLD, cirrhosis) with consequent failures of the liver, kidneys, circulation, lungs, and hemostasis; all of them were diagnosed by CLIF-modified SOFA scores, nowadays available as an internet calculator. Similarly determined by the study results, was the characteristically high short-term mortality of ACLF. Subsequent studies, although heterogeneous due to the differences of the diagnostic criteria of Europe, USA, and Asia, brought about the proof-of-concept with additional important findings on the dynamics of the syndrome, during the first days of clinical presentation, as well as the close pathophysiological link between ACLF, immunity, inflammation, and infections. The background immunological milieu of ACLD is that of immune deficit (CAIDS), gut dysbiosis, leaky gut, and chronic systemic low-grade inflammation. Most of the hits (triggers), which use to convert ACLD to ACLF are inflammatory in nature, as is the response of organism. The prevailing immunological status truncate prognosis into fulminant death, early recovery, recovery due to intervention in the golden window, the catabolic draw of PICS, and indolent death. Although the therapeutic interventions are mostly non-specific, their intensive implementation can lead to improvement in the survival in all the categories of ACLF. Immunity, inflammation, and infections are in the focus of current scientific interest; ligands and receptors of known pathways, gut microbiome, gut barrier, and bacteria are the main therapeutic targets. Liver regeneration, liver replacement, and futility are another hot topics of ACLF research. Until its results become available, the cornerstones of management of ACLF will remain based on the adherence to the basic principles – early diagnosis, management on ICU, emphasis on infections and inflammation, early prognostic re-stratification, and previously tough decisions about liver replacement/regeneration, and futility. Key words: acute-over-chronic liver failure, immunity, inflammation, infection, management.

Address for correspondence Lubomir Skladany, MD, PhD, Head, Department of Internal Medicine II, Slovak Medical University, F.D. Roosevelt University Hospital, Nam. L. Svobodu 1, 97401 Banská Bystrica, e-mail: [email protected]

Introduction The syndrome named ACLF (acute-over-chronic liver failure) has relatively short history. In 1995, it was mentioned by Ohnishi; in 2002, a  dedicated working party was organized in London; in 2009, Chronic Liver Failure (CLIF) Consortium was created, whose endeavor led to the landmark study on ACLF in 2013. The CANONIC study set the scene for the introduction of ACLF to clinical practice [1]. Ever since, CLIF Consortium, European Association for the Study of the Liver (EASL) (jointly EASL-CLIF), Asian Pacific

Clinical and Experimental Hepatology Suppl 1/2017

Association for the Study of the Liver (APASL), North American Consortium for the Study of End-Stage Liver Disease (NACSELD), and several other bodies have striven for unifying diagnostic criteria, which were consented in 2014 [1-7]. The main question fueling the debate around the diagnostic criteria has been the necessity (or not) of the clear-cut liver cirrhosis as the background stage of the underlying liver disease. This problem was indirectly mitigated by the Baveno VI recommendation to call the term ‘advanced chronic liver disease’ (ACLD), rather than cirrhosis [5, 8]. From the 15% new decompensations per year of previously

s7

Lubomir Skladany, Svetlana Adamcova-Selcanova, Daniela Jancekova

asymptomatic ACLD, ACLF was present in around 25% at admission, and developed in another 10% during hospitalization; therefore, ACLF can be considered a common entity [1, 4, 9-14]. At least in the medical environment proximate to the authors, there has been noticeable hesitation to accept ACLF as a self-existing entity. One may and probably should ask: why? One explanation could be that the abyss lying between the toll ACLF claims on the one side, and the lack of disease-specific therapy on the other, creates environment of hopelessness and frustration, leading to evasive behavior. This causal cascade, operative in the ACLF arena for years, has been accompanied by the increasing money-sensitivity of medical care: the almost prohibitive cost per life saved could be but understandable hindrance. The cost at the intensive care unit (ICU) can be as high as 50,000 £, one hospitalization could reach up to 116-180 $, and one year expenditures – 3 billion $ [15, 16]. The admission of patient with acute decompensation (AD) to ICU means green-light for giving out the money. On the other hand, turning blind eye means nobody would care, because patients with decompensated (d)ACLD simply use to die, and there is no specific therapy for possible ACLF anyway. However, this standpoint cannot be accepted anymore. First, there is an ontological reason. Only acceptance of the limitations could lead to the brainstorming necessary for progress. Two studies have already shown how mere admission to the ICU has led to decrease in mortality and improvement of survival [17, 18]; the new pathophysiological paradigm – ‘from peripheral arterial vasodilation to systemic inflammation hypothesis’ – has been formulated [19], and new treatment options are on the horizon (see below).

Decompensating events « Ascites « Jaundice « Variceal bleeding « Hepatic encephalopathy Chronic liver disease Precipitants « Alcohol « Sepsis « Virus « Drug « Ischemic « Surgery « Idiopathic

Compensated cirrhosis

Decompensated cirrhosis

Definition In the CANONIC study, ACLF was defined as AD of liver cirrhosis associated with organ/system failure(s), which may develop any time during the course of the disease, and is associated to a short-term (28-day) mortality, ranging from 23% to 74%, depending on the number of organ failures [1]. Currently, ACLF is defined as an acute deterioration of preexisting, chronic liver disease (with or without the presence of liver cirrhosis), due to a  precipitating event with additional failure of one or more organs or systems, and associated with high short-term mortality [7, 10]. Despite evidence-based definition and diagnostic criteria, ACLF population remains heterogenous [20].

Timeframe The first step to the diagnosis of ACLF in patients with dACLD is the determination of its duration: ACLF should be suspected in each patient with AD, defined as the appearance of the decompensating event 2-4 weeks before admission [20]. Because ACLF is a very dynamic process, it is recommended to re-calculate its grade once more anytime 3 to 7 days after the diagnosis, since at this point, 81% of patients achieve their final ACLF grade, and this grade predicts mortality better than that at admission (see below) [21, 22].

Decompensating events (Fig. 1) All the so-called specific complications of ACLD are to be considered the potential decompensating events, by which ACLF presents itself: i.e., ascites, jaundice, portal hypertensive bleeding, hepatic encephalopathy (HE), and bacterial infections. If the first ever decompensating event is full-blown ACLF, the mortality is higher than in patients with the history of previous decompensations [1]. The role of the etiology of underlying ACLD was the focus of interest in at least eight studies, summarized by Abbas et al. [23, 24].

Diagnostic criteria CLIF C score Type A ACLF

Type B ACLF

Hepatic and extrahepatic organ failures

Fig. 1. Subtypes of acute-over-chronic liver failure (ACLF) [5]

s8

Diagnosis and prognosis

Type C ACLF

Diagnostic criteria are based on three domains: 1) AD of 2) ACLD with 3) organ/system failure(s). Currently, provided the presence of clinical suspicion, diagnosis of ACLF is easy: all the patients admitted to the hospital with AD (timeframe + decompensating events) should be examined for the presence of ACLF by uploading their clinical and laboratory variables into

Clinical and Experimental Hepatology Suppl 1/2017

Acute-over-chronic liver failure (ACLF) – immunity, infection, inflammation

Table 1. CLIF-OF scoring system for diagnosing acute over chronic liver failure (ACLF) [5] Organ system

Score = 1

Score = 2

Score = 3

Liver: bilirubin (mg/dl)

< 6.0

≤ 6.0-12.0

> 12.0

Kidney: creatinine (mg/dl)

< 2.0

so%

sX[[NWJU[NYUJLNVNW]]QN[JYb

Cerebral: HE grade

Grade 0

Grade 1-2

Grade 3-4

Coagulation: INR

2.0-2.5

s

Circulation: MAP (mmHg)

s VV1P

< 70 mmHg

Vasopressors

Respiratory: PaO2/FiO2 or SpO2/FiO2)

> 300 > 357

> 200 – ≤ 300 > 214 – ≤ 357

≤ 200 ≤ 214

HE – hepatic encephalopathy, INR – international normalised ratio, MAP – mean arterial pressure, PaO2 – arterial partial pressure of oxygen, SpO2 – oxygen saturation, FiO2 – fraction of inspired oxygen

Table 2. Acute over chronic liver failure (ACLF) grades and prognosis [86]

28-day and 90-day mortality rate (%)

ACLF grade 1

1) No organ failure 2) Single organ failure (liver, coagulation, circulation, lungs) in patients with serum creatinine levels < 133 mmol/l and no hepatic encephalopathy 3) Single cerebral failure in patients with serum creatinine levels < 133 mmol/l

1) Single kidney failure 2) Single organ failure in patients with serum creatinine levels ranging from 133 mmol/l to 176 mmol/l and/or grade 1-2 hepatic encephalopathy 3) Single cerebral failure with serum creatinine from 133 mmol/l to 176 mmol/l

4.7; 14.0

22.1; 40.7

the freely available web-calculator (www.clifresearch. com/ToolsCalculators.aspx) [11]. As mentioned, coining the term ACLD instead of cirrhosis also contributed to simplification of the diagnostic process [8]. i) The original (West) diagnostic criteria issued by the CANONIC study were based on the presence of liver cirrhosis, AD, and organ/system failures determined by CLIF-modified Sequential Organ Failure Score (CLIF-SOFA) [1] (Table 1). ii) The APASL (East) criteria have broadened the cohort by the (abundant in the East) patients without liver cirrhosis, and excerpted the hepatic from the insults, as well as ascites and HE from events [2, 3]. iii) Unifying consensus definition resolved some of the issues by dividing the syndrome to types A-C (Fig. 1) [5]. The quantitative influence on mortality of all the individual organ-, and system-failures included in CLIF-SOFA scoring system, i.e., the liver, coagulation, kidney, circulation, brain, and lung, was subsequently revised [25]. Therefore, patients with single organ failure are not automatically included to ACLF 1; rather, according to the organ involved, they were divided into ACLF 1a, and 1b (Table 2). Apart from confirming the presence (or absence) of the ACLF, web calculator also displays the probability of 28-day mortality (Fig. 2) and, after uploading the

Clinical and Experimental Hepatology Suppl 1/2017

ACLF grade 2

ACLF grade 3

Defined by the presence Defined by the presence of two organ failures of three organ failures or more

32.0; 52.3

76.7; 79.1

age and white blood cells count (WBC), the calculator offers additional refinement of prognosis in patients with AD.

Precipitating events Precipitating events (insults, triggers) are detectable in two-thirds to 50% of cases [1]. The knowledge of their local representation allows for the comparison of cohorts and built-up of prevention [26]. On the other hand, knowing the precipitating event of an in90 80 70 Mortality rate (%)

Definitions ACLF

No ACLF

60 50 40 30 20 10 0

NO ACLF

ACLF 1 28-day mortality

ACLF 2 90-day mortality

ACLF 3

Fig. 2. Survival according to the acute over chronic liver failure (ACLF) grade [1]

s9

Lubomir Skladany, Svetlana Adamcova-Selcanova, Daniela Jancekova

dividual patient, further refines his or her prognostic stratification and enables personalized approach; intrahepatic events bear poorer prognosis than extrahepatic [13].Therefore, the division of events to extrahepatic (bleeding, infection, surgery etc.) and intrahepatic (e.g., flares of chronic hepatitis B, autoimmune hepatitis, Wilson’s disease; superinfection by hepatitis A, or hepatitis E virus infection, acute alcoholic hepatitis, drug-induced liver injury, etc.), is of prognostic importance. Of note, large-volume paracentesis without albumin replacement is one of the listed triggers [24]. In Western countries, ACLF usually occurs in a context of systemic inflammatory response syndrome (SIRS) due to severe alcoholic hepatitis, bacterial infection, or to unknown causes suspected of bearing the relation to the gut-liver axis [1, 4].

Time-course One of the hallmarks of ACLF is its dynamic nature; the changes (for better or worse) in ACLF grades have occurred very rapidly, rapidly, or slowly in 40%, 15%, and 15% of the patients, respectively [21]. The evolution of the ACLF grades between the two time-points (1 – at admission, 2 – at any time from day 3 to day 7), gives the additional prognostic information over the ACLF grade at admission; the ACLF grade at the 2nd time-point has been termed ‘final ACLF’ [21]. The 28-day mortality of the final ACLF1 has been worse than that of the resolved ACLF (18% vs. 6%), but distinctively better than that of the final ACLF 2-3 (42-92%) [21]. This lends support to the case for the so-called ‘golden window’, or ‘window of opportunity’ (for the intervention to be most effective) coined by Sarin [27, 28]. The resolution of ACLF could be achieved in around 40% (53%, 35%, and 16% for the ACLF grades 1, 2, and 3, respectively), and was accompanied by low short-term mortality (6%), as compared to the unresolved ACLF (18%) [21]. Another consequence of the first-week dynamics is the subdivision of ACLF to the severe early course (final ACLF > 1), and non-severe early course (final ACLF ≤ 1) [21]. The severe early course cohort seems to be sufficiently homogenous and enriched for the study of therapeutic measures [29, 30]. Half the patients with severe early course were progressed from ACLF 1, 45% were non-movers, and 5% were improvers from ACLF 3 to ACLF 2 [21].

Inflammation and ACLF Whereas the liver has tendency to establish immune tolerance, one of the most prominent features of ACLF has been the escalated systemic inflammation

s10

[24, 31]. The situation is further complicated by the fact that ACLF is evolving in the setting of chronic immune dysfunction and low-grade inflammation of ACLD, known as cirrhosis-associated immune dysfunction syndrome (CAIDS) [32]. It was the very strength of the association between the degree of liver injury and immune deficit, and relatively standard structure of immune derangements in CAIDS, which reduced infections specific complications of ACLD [28, 33]. Pro-inflammatory state (SIRS) of ACLF can itself be robust enough to kill [34] (Fig. 3). Ignited mainly by the early innate immune response, SIRS could be the dominant pathophysiological basis of the multiorgan failure (MOF) and death, termed in this case as ‘fulminant death’ (Fig. 3) [35]. Subsequently, SIRS is counterbalanced by compensatory anti-inflammatory response syndrome (CARS) [36]. The main purpose of CARS is beneficial – to prevent autogenous damage to the organs and tissues by the excessive inflammation [36]. To quote from the Rosenthal et al. “...while SIRS was a pro-inflammatory syndrome that seemed tasked with killing infectious organisms through activation of the immune system, CARS was a  systemic deactivation of the immune system tasked with restoring homeostasis from an inflammatory state” [35]. There are two situations, however, in which CARS can turn detrimental: if its intensity overbalances SIRS in such that the result is the so-called immune paralysis, or if the length of duration of SIRS-CARS co-existence leads to the exhaustion of immune and energy reserves with catabolic state called persistent inflammatory, immunosuppressed, catabolic syndrome (PICS) [35]. Some authors call these patients chronically critically ill [37]. One way or another, CARS is setting the stage for late nosocomial infections that can precipitate late-onset MOF, and the so-called ‘indolent death’ (Fig. 3) [36, 38, 39]. Molecular and cellular mechanisms behind CAIDS, SIRS, CARS, and PICS were described elsewhere [34-36, 38-41]. There are two inflammatory contexts of ACLF: with and without infection [24]. The first, infection-associated, represents at least 30% of the burden, and has been designated (by NACSELD) as iACLF [4, 42-44]. Infection was the offence in 38-80% of all ACLF cases [1, 11, 13], and even more significant if they were of nosocomial origin [12]. In submitted study by Fernandez et al., referred at the International Liver Congress 2017, 25% and 37% of patients with AD and ACLF had infection at admission, respectively. In non-infected patients from the same study, 18%, 37%, 47%, and 81% acquired bacterial infections during the course of AD, ACLF 1, ACLF 2, and ACLF 3, respectively. That ACLF is the risk factor for acquisition of infections – with

Clinical and Experimental Hepatology Suppl 1/2017

Acute-over-chronic liver failure (ACLF) – immunity, infection, inflammation

Immune stimulation Pro-inflammatory processes

Fulminant death

MODS

GOLDEN WINDOW

Persistent SIRS

S SIR HIT

Immunosuppression Anti-inflammatory processes

CAIDS

EARLY RECOVERY

LATE RECOVERY

CA R

Time (days 0-28)

S

PICS

Persiste

nt CARS

Im mu

ne

par

aly s

is Indolent death

PICS – persistent inflammatory, immunosuppressed, catabolic syndrome, MOF – multiple organ failure, SIRS – systemic inflammatory response syndrome, CARS – compensatory antiinflammatory response syndrome

Fig. 3. Acute over chronic liver failure (ACLF) and inflammatory responses [35]

the frequency twice as high as in chronic liver failure (58% vs. 26%, p = 0.007) was shown in the study by Katoonizadeh [45]. Bacteria trigger inflammation by their PAMPs (pattern associated molecular patterns), or by their virulence factors, both of which engage PRRs (pattern recognition receptors), and launch the so-called structural feature recognition [24]. The most well-known PRRs is toll-like receptor (TLR) family, present on the innate-immunity and epithelial cells. Receptors for ligands of other microorganisms, such as viruses and fungi, are different from those of bacteria. Mortality in bacterial sepsis is higher in patients with cirrhosis than without, and the second infection in one ACLF multiplies the mortality [43, 46]. The inflammatory phenotype of ACLF without infection is characterized by elevated levels of proinflammatory markers such as WBC, C-reactive protein, interleukin (IL)-6, IL-1β, and IL-8 [41, 47, 48]. The prototype of non-infectious inflammatory ACLF phenotype is acute alcoholic hepatitis. Inflammation in non-infectious context is triggered by the released content of damaged cells, or broken-down extracellular matrix [24, 49-51]. Together, they are known as danger-associated molecular patterns (DAMPs), and are – with their respective receptors – listed in the review by Hernaez et al. [24]. If the trigger of inflammation is not identifiable, three possible sources are postulated: changed microbiota with translocated metabolites of gut bacteria, translocated PAMPs of gut bacteria, and DAMPs [4, 52-54].

Clinical and Experimental Hepatology Suppl 1/2017

Management There is no specific treatment of ACLF available [55, 56]. As already mentioned, mortality can be decreased by admitting all the ACLF patients to the ICU, presumed cost notwithstanding. It is important to strive to hit the golden window, the timeframe of opportunity for SIRS-CARS tension to be lessened before one of them will prevail in either fulminant or indolent death; or, before their prolonged draw will cause PICS. In fact, they are exactly patients with PICS who drain the most of the resources, with enthusiasm for ACLF included. The basic principles of the management of ACLF could be summarized as follows: 1. Early diagnosis by extracting AD from dACLD, uploading variables to clifresearch.com. 2. Prognostic stratification of ACLF at days 0 and 7 with the a) final ACLF, and b) non/severe early course as the bases for decisionmaking. 3. Intensive care and clinical nutrition, aimed at the improvement of organ failures present at the diagnosis, and prevention of the impairment of other organs during hospitalization [9, 57]. 4. Combatting bacterial infections by hitting fast and hard in the empirical interval by choosing the most powerful, broadest-spectrum antibiotics according to the actual hospital epidemiology/microbiology results [58]. In the long-term, the cornerstones of

s11

Lubomir Skladany, Svetlana Adamcova-Selcanova, Daniela Jancekova

success is the evidence-based antibiotic stewardship [59, 60]. If the insult is new-onset (e.g., hepatitis E) or flare-up (e.g., hepatitis B) of viral hepatitis, the therapy is straight forward. 5. Manipulation of the inflammation and immunity. Stimulation of the immunity has been investigated by some groups based on the premise that highgrade SIRS notwithstanding, the prevailing immune milieu during the golden window has been immune deficit, resulting from the combination of background CAIDS with CARS [41]. Growth factors for WBC (G-CSF) in 12-dose protocol, erythropoietin (in combination with G-CSF), or other non-specific immune stimulants such as transfer factor, has been all tested in pilot studies with promising results [61-64]. It has to be pointed out that both G-CFM and EPO were also perceived as the mediators of regeneration for hepatocytes [65, 66]. Microbiome and inflammasomes are another targets for immune manipulation, with rifaximin and some other antibiotics and drugs being studied as potential disease- modifying anti-cirrhotic drugs (DMACT) [67-71]. Anti-inflammatory measures are to be tested in the search for DMACTs (P. Caraceni, ILC® 2017). Solving their seemingly oxymoronous relationship with the immune-stimulating approaches is the area of intensive research [72]. One of the examples could be the treatment of acute alcoholic hepatitis [73-75]. 6. ‘Liver failure is the failure of liver regeneration’ said the journal ‘Science’ in 1997. Suboptimal regeneration of hepatocytes is therapeutic target distilled from the deepening knowledge of the pathophysiology of liver failure, and ACLF [62, 76-78]. New hepatocytes are derived from the liver- or bone-marrow niches, respectively [79, 80]. Growth factors (G-CFM, EPO) mentioned above are one of possible mediators of regeneration. There were at least six and four studies on the augmentation of the liver regeneration, by the sorted and unsorted bone marrow-derived mononuclear cells, respectively [81]. Other possibilities are the hepatocyte transplantation and bioartificial liver. 7. Fast-track liver transplantation (LTx) in severe early course, especially in non-responders to therapy of ACLF, so-called salvage LTx, is the area of intensive study [82-85]. Since the prognosis of the severe early course ACLF is dismal (28-day mortality, 34-51%), LTx (1-year survival > 75%), aroused as the logical area to investigate. However, the uptakes of LTx in pivotal trials were only 2.2%, and 4.9%, and the waiting-list mortality more than 50% [21, 84, 85]. The prioritization on the waiting list (the sickest

s12

first?), timing of LTx (the sooner the better?), futility (too sick to be transplanted?), and net transplant benefit (the good for all?) are the areas of further research in ACLF [86, 87]. As for the too sick to be transplanted, proposed for confirmatory studies are ACLF points > 64, 3 organ supports, ACLF 3, respiratory failure, intensity of organ support (e.g., vasopressor dose), and the duration of organ support [88]. 8. Liver support. Whereas general intensive care focuses on the support of the other organs and systems, liver support deals with the core organ of ACLF – the liver. So-called extracorporeal liver support systems (MARS, Prometheus) are the only more widely available modalities from this group, which were studied in two tens of dozens of patients with ACLF; none of the methods led to the improvement of overall survival [29, 30]. 9. Never give up. Until the new studies will clearly show otherwise, or until informed and competent patient requests quitting AND the family requests to quit, AND the interdisciplinary team unequivocally consents that the probability of recovery is negligible, there is no other option than to continue therapy [21]. Studies in this area are underway.

Conclusions ACLF is the syndrome with significant prevalence in patients admitted to the hospital with dACLD with distinct diagnostic features and high short-term mortality. It is worth uploading the few variables of the patients with AD to the web calculator and see if the criteria for ACLF are fulfilled. If they are, the patient should be admitted to the ICU. Since the core pathophysiology of the syndrome has much to do with immunity, inflammation, and infections, the most specific of current non-specific measures are directed their way, as is the research. Salvage LTx is the evidence-based option, but lot of issues remain to be resolved; who is too sick to be transplanted, and whose liver parenchyma could be helped to regenerate over the critical mass, are but the few. References 1. Moreau R, Gines P, Jalan R, et al. Acute-on-chronic liver failure is a distinct syndrome that develops in patients with acute decompensation of cirrhosis. Gastroenterol 2013; 144: 1426-1437. 2. Sarin SK, Kumar A, Almeida JA, et al. Acute-on-chronic liver failure: consensus recommendations of the Asian Pacific Association for the Study of the Liver (APASL). Hepatol Int 2009; 3: 269-282. 3. Sarin SK, Kedarisetty CK, Abbas Z, et al. Acute-on-chronic liver failure: consensus recommendations of the Asian Pacific Asso-

Clinical and Experimental Hepatology Suppl 1/2017

Acute-over-chronic liver failure (ACLF) – immunity, infection, inflammation

4. 5. 6. 7. 8.

9. 10.

11.

12.

13. 14. 15. 16. 17. 18.

19.

20. 21.

22.

ciation for the Study of the Liver (APASL) 2014. Hepatol Int 2014; 8: 453-471. Bajaj JS, O’Leary JG, Reddy KR, et al. Survival in infection-related acute-on-chronic liver failure is defined by extrahepatic organ failures. Hepatology 2014; 60: 250-256. Jalan R, Yurdaydin C, Bajaj JS, et al. Toward an improved definition of acute-on-chronic liver failure. Gastroenterology 2014; 147: 4-10. Jalan R, Fernandez J, Wiest R, et al. Bacterial infections in cirrhosis: a position statement based on the EASL Special Conference 2013. J Hepatol 2014; 60: 1310-1324. Jalan R, Saliba F, Pavesi M, et al. Development and validation of a prognostic score to predict mortality in patients with acuteon-chronic liver failure. J Hepatol 2014; 61: 1038-1047. De Franchis R; Baveno VI Faculty. Expanding consensus in portal hypertension: Report of the Baveno VI Consensus Workshop: Stratifying risk and individualizing care for portal hypertension. J Hepatol 2015; 63: 743-752. Bernal W, Jalan R, Quaglia A, et al. Acute-on-chronic liver failure. Lancet 2015; 386: 1576-1587. Jalan R, Pavesi M, Saliba F, et al. The CLIF Consortium acute decompensation score (CLIF-C ADs) for prognosis of hospitalized cirrhotic patients without acute-on-chronic liver failure. J Hepatol 2015; 62: 831-840. Arroyo V, Moreau R, Jalan R, et al.; EASL-CLIF Consortium CANONIC Study. Acute-on-chronic liver failure: A  new syndrome that will re-classify cirrhosis. J Hepatol 2015; 62 (1 Suppl): S131-143. Sargenti K, Prytz H, Nilsson E, et al. Predictors of mortality among patients with compensated and decompensated liver cirrhosis: the role of bacterial infections and infection-related acute-on-chronic liver failure. Scand J Gastroenterol 2015; 50: 875-883. Shi Y, Yang Y, Hu Y, et al. Acute-on-chronic liver failure precipitated by hepatic injury is distinct from that precipitated by extrahepatic insults. Hepatology 2015; 62: 232-242. Li H, Chen LY, Zhang NN, et al. Characteristics, diagnosis and prognosis of acute-on-chronic liver failure in cirrhosis associated to hepatitis B. Sci Rep 2016; 6: 25487. Shawcross DL, Austin MJ, Abeles RD, et al. The impact of organ dysfunction in cirrhosis: survival at a cost? J Hepatol 2012; 56: 1054-1062. Olson JC, Wendon JA, Kramer DJ, et al. Intensive care of the patient with cirrhosis. Hepatology 2011; 54: 1864-1872. Schmidt M, Barritt A, Orman E, et al. Decreasing mortality among patients hospitalised with cirrhosis in the United States from 2002 through 2010. Gastroenterology 2015; 148: 967-977. McPhail MJ, Shawcross DL, Abeles RD, et al. Increased survival for patients with cirrhosis and organ failure in liver intensive care and validation of the chronic liver failure-sequential organ failure scoring system. Clin Gastroenterol Hepatol 2015; 13: 1353-1360. Bernardi M, Moreau R, Angeli P, et al. Mechanisms of decompensation and organ failure in cirrhosis: From peripheral arterial vasodilation to systemic inflammation hypothesis. J Hepatol 2015; 63: 1272-1284. Moreau R, Arroyo V. Acute-on-chronic liver failure: a new clinical entity. Clin Gastroenterol Hepatol 2015; 13: 836-841. Gustot T, Fernandez J, Garcia E and the CANONIC Study Investigators of the EASL-CLIF Consortium. Clinical course of acute-on-chronic liver failure syndrome and effects on prognosis. Hepatology 2015; 62: 243-252. Jalan R, Stadlbauer V, Sen S, et al. Role of predisposition, injury, response and organ failure in the prognosis of patients with

Clinical and Experimental Hepatology Suppl 1/2017

23. 24. 25. 26. 27. 28. 29.

30.

31. 32. 33. 34.

35. 36. 37. 38. 39.

40.

41.

42.

acute-on-chronic liver failure: a prospective cohort study. Crit Care 2012; 16: R227. Abbas Z, Shazi L. Pattern and profile of chronic liver disease in acute on chronic liver failure. Hepatol Int 2015; 9: 366-372. Hernaez R, Solà E, Moreau R, et al. Acute-on-chronic liver failure: an update. Gut 2017; doi: 10.1136/gutjnl-2016-312670 [Epub ahead of print]. Wlodzimirow KA, Eslami S, Abu-Hanna A, et al. A systematic review on prognostic indicators of acute on chronic liver failure and their predictive value for mortality. Liver Int 2013; 33: 40-52. Bajaj JS. Defining acute-on-chronic liver failure: will East and West ever meet? Gastroenterology 2013; 144: 1337-1339. Shaw AC, Goldstein DR, Montgomery RR. Age-dependent dysregulation of innate immunity. Nat Rev Immunol 2013; 13: 875-887. Sarin SK, Choudhury A. Acute-on-chronic liver failure: terminology, mechanisms and management. Nat Rev Gastroenterol Hepatol 2016; 13: 131-149. Kribben A, Gerken G, Haag S, et al. Effects of fractionated plasma separation and adsorption on survival in patients with acute-on-chronic liver failure. Gastroenterology 2012; 142: 782-789. Bañares R, Nevens F, Larsen FS and the RELIEF study group. Extracorporeal albumin dialysis with the molecular adsorbent recirculating system in acute-on-chronic liver failure: the RELIEF trial. Hepatology 2013; 57: 1153-1162. Crispe IN. The liver as a lymphoid organ. Annu Rev Immunol 2009; 27: 147-163. Bonnel AR, Bunchorntavakul C, Reddy KR. Immune dysfunction and infections in patients with cirrhosis. Clin Gastroenterol Hepatol 2011; 9: 727-738. Chovatiya R, Medzhitov R. Stress, inflammation, and defense of homeostasis. Mol Cell 2014; 54: 281-288. Bone RC, Balk RA, Cerra FB, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest 1992; 101: 1644-1655. Rosenthal MD, Moore FA. Persistent inflammatory, immunosuppressed, catabolic syndrome (PICS): A  new phenotype of multiple organ failure. J Adv Nutr Hum Metab 2015; 1: e784. Bone RC. Sir Isaac Newton, sepsis, SIRS, and CARS. Crit Care Med 1996; 24: 1125-1128. Nierman DM. A structure of care for the chronically critically ill. Crit Care Clin 2002; 18: 477-491. Ward NS, Casserly B, Ayala A. The compensatory anti-inflammatory response syndrome (CARS) in critically ill patients. Clin Chest Med 2008; 29: 617-625. Gentile LF, Cuenca AG, Efron PA, et al. Persistent inflammation and immunosuppression: a common syndrome and new horizon for surgical intensive care. J Trauma Acute Care Surg 2012; 72: 1491-1501. Basta G, Del Turco S, Navarra T, et al.; Acute Liver Failure Study Group. Circulating levels of soluble receptor for advanced glycation end products and ligands of the receptor for advanced glycation end products in patients with acute liver failure. Liver Transpl 2015; 21: 847-854. Bernsmeier C, Pop OT, Singanayagam A, et al. Patients with acute-on-chronic liver failure have increased numbers of regulatory immune cells expressing the receptor tyrosine kinase MERTK. Gastroenterology 2015; 148: 603-615. Gustot T, Felleiter P, Pickkers P, and the EPIC II group of investigators. Impact of infection on the prognosis of critically ill

s13

Lubomir Skladany, Svetlana Adamcova-Selcanova, Daniela Jancekova

43. 44.

45. 46.

47. 48. 49. 50. 51.

52. 53. 54. 55. 56. 57.

58. 59. 60. 61. 62. 63.

s14

cirrhotic patients: results from a  large worldwide study. Liver Int 2014; 34: 1496-1503. Gustot T, Durand F, Lebrec D, et al. Severe sepsis in cirrhosis. Hepatology 2009; 50: 2022-2033. Arvaniti V, D‘Amico G, Fede G, et al. Infections in patients with cirrhosis increase mortality four-fold and should be used in determining prognosis. Gastroenterology 2010; 139: 1246-1256, 1256.e1-5. Katoonizadeh A, Laleman W, Verslype C, et al. Early features of acute-on-chronic alcoholic liver failure: a prospective cohort study. Gut 2010; 59: 1561-1569. Bajaj JS, O’Leary JG, Reddy KR, et al.; NACSELD. Second infections independently increase mortality in hospitalized patients with cirrhosis: the North American consortium for the study of end-stage liver disease (NACSELD) experience. Hepatology 2012; 56: 2328-2335. Claria J, Stauber R, Coenraad MJ, et al. Systemic Inflammation in decompensated cirrhosis. Characterization and role in acute on chronic liver failure. Hepatology 2016; 64: 1249-1264. Sole C, Sola E, Morales-Ruiz M, et al. Systemic inflammatory response profile in acute-on-chronic liver failure and its relationship with prognosis. Sci Rep 2016; 6: 32341. Medzhitov R. Origin and physiological roles of inflammation. Nature 2008; 454: 428-435. Kono H, Rock KL. How dying cells alert the immune system to danger. Nat Rev Immunol 2008; 8: 279-289. Wang X, Sun R, Wei H, et al. High-mobility group box 1 (HMGB1)Toll-like receptor (TLR)4-interleukin (IL)-23-IL-17A axis in druginduced damage-associated lethal hepatitis: Interaction of γδ T cells with macrophages. Hepatology 2013; 57: 373-384. Arroyo V, Moreau R, Kamath PS, et al. Acute-on-chronic liver failure in cirrhosis. Nat Rev Dis Primers 2016; 2: 16041. Qin N, Yang F, Li A, et al. Alterations of the human gut microbiome in liver cirrhosis. Nature 2014; 513: 59-64. Bajaj JS, Betrapally NS, Gillevet PM. Decompensated cirrhosis and microbiome interpretation. Nature 2015; 525: E1-2. Gines P, Fernandez J, Durand F, et al. Management of criticallyill cirrhotic patients. J Hepatol 2012; 56 (Suppl 1): S13-24. Nadim MK, Durand F, Kellum JA, et al. Management of the critically ill patient with cirrhosis: a multidisciplinary perspective. J Hepatol 2016; 64: 717-735. Angeli P, Gines P, Wong F, et al.; International Club of Ascites. Diagnosis and management of acute kidney injury in patients with cirrhosis: revised consensus recommendations of the International Club of Ascites. Gut 2015; 64: 531-537. Arabi YM, Dara SI, Memish Z, et al. Antimicrobial therapeutic determinants of outcomes from septic shock among patients with cirrhosis. Hepatology 2012; 56: 2305-2315. Ge PS, Runyon BA. Preventing future infections in cirrhosis: a battle cry for stewardship. Clin Gastroenterol Hepatol 2015; 13: 760-762. Šváč J, Skladaný L, Purgelová A, et al. Antimicrobial stewardship program (ASP) on hepatogastrotransplantation unit (HEGITO) [In press]. Singh V, Sharma AK, Narasimhan RL, et al. Granulocyte colony-stimulating factor in severe alcoholic hepatitis: a randomized pilot study. Am J Gastroenterol 2014; 109: 1417-1423. Moreau R, Rautou PE. G-CSF therapy for severe alcoholic hepatitis: targeting liver regeneration or neutrophil function? Am J Gastroenterol 2014; 109: 1424-1426. Duan XZ, Liu FF, Tong JJ, et al. Granulocyte-colony stimulating factor therapy improves survival in patients with hepatitis B virus-associated acute-on-chronic liver failure. World J Gastroenterol 2013; 19: 1104-1110.

64. Kedarisetty CK, Anand L, Bhardwaj A, et al. Combination of granulocyte colony-stimulating factor and erythropoietin improves outcomes of patients with decompensated cirrhosis. Gastroenterology 2015; 148: 1362-1370. 65. Garg H, Kumar A, Sharma BC, et al. Clinical profile and predictors of mortality in patients of acute on chronic liver failure. Dig Liver Dis 2012; 44: 166-171. 66. Khanam A, Trehanpati N, Sharma BC, et al. Altered frequencies of dendritic cells and IFN-gamma-secreting T cells with granulocyte colony-stimulating factor (G-CSF) therapy in acute-onchronic liver failure. Liver Int 2014; 34: 505-513. 67. Chassaing B, Etienne-Mesmin L, Gewirtz AT. Microbiota-liver axis in hepatic disease. Hepatology 2014; 59: 328-339. 68. Zaki MH, Lamkanfi M, Kanneganti TD. The Nlrp3 inflammasome: contributions to intestinal homeostasis. Trends Immunol 2011; 32: 171-179. 69. Henao-Mejia J, Elinav E, Jin C, et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012; 482: 179-185. 70. Kakiyama G, Pandak WM, Gillevet PM, et al. Modulation of the fecal bile acid profile by gut microbiota in cirrhosis. J Hepatol 2013; 58: 949-955. 71. Dhiman RK, Agrawal S, Gupta T, et al. Chronic Liver Failure-Sequential Organ Failure Assessment is better than the Asia-Pacific Association for the Study of Liver criteria for defining acute-on-chronic liver failure and predicting outcome. World J Gastroenterol 2014; 20: 14934-14941. 72. Selvapatt N, Singanayagam A, Wendon J, et al. Understanding infection susceptibility in patients with acute-on-chronic liver failure. Intensive Care Med 2014; 40: 1363-1366. 73. Thursz MR, Richardson P, Allison M, et al.; STOPAH Trial. Prednisolone or pentoxifylline for alcoholic hepatitis. N Engl J Med 2015; 372: 1619-1628. 74. Nguyen-Khac E, Thevenot T, Piquet MA, et al.; AAH-NAC Study Group. Glucocorticoids plus N-acetylcysteine in severe alcoholic hepatitis. N Engl J Med 2011; 365: 1781-1789. 75. Mathurin P, Bataller R. Trends in the management and burden of alcoholic liver disease. J Hepatol 2015; 62 (1 Suppl): S38-46. 76. Dubuquoy L, Louvet A, Lassailly G, et al. Progenitor cell expansion and impaired hepatocyte regeneration in explanted livers from alcoholic hepatitis. Gut 2015; 64: 1949-1960. 77. Tygstrup N. Aspects of hepatic hypoxia: observations on the isolated, perfused pig liver. Bull N Y Acad Med 1975; 51: 551-556. 78. Ranek L. Cytophotometric studies of the DNA, nucleic acid and protein content of human liver cell nuclei. Acta Cytol 1976; 20: 151-157. 79. Maiwall R, Kumar A, Sarin SK. Liver regeneration during acuteon-chronic liver failure using growth factors: in vivo or ex vivo indulgence of bone marrow? Gastroenterology 2013; 145: 901-904. 80. Bihari C, Anand L, Rooge S, et al. Bone marrow stem cells and their niche components are adversely affected in advanced cirrhosis of the liver. Hepatology 2016; 64: 1273-1288. 81. Shasthry SM, Sarin SK. New treatment options for alcoholic hepatitis. World J Gastroenterol 2016; 22: 3892-3906. 82. Pereira MR, Scully BF, Pouch SM, et al. Risk factors and outcomes of carbapenem-resistant Klebsiella pneumoniae infections in liver transplant recipients. Liver Transpl 2015; 21: 1511-1519. 83. Bahirwani R, Shaked O, Bewtra M, et al. Acute-on-chronic liver failure before liver transplantation: impact on posttransplant outcomes. Transplantation 2011; 92: 952-957. 84. Finkenstedt A, Nachbaur K, Zoller H, et al. Acute-on-chronic liver failure: excellent outcomes after liver transplantation but high mortality on the wait list. Liver Transpl 2013; 19: 879-886.

Clinical and Experimental Hepatology Suppl 1/2017

Acute-over-chronic liver failure (ACLF) – immunity, infection, inflammation

85. Reddy KR, O‘Leary JG, Kamath PS, et al.; North American Consortium for the Study of End-Stage Liver Disease. High risk of delisting or death in liver transplant candidates following infections: Results from the North American Consortium for the Study of End-Stage Liver Disease. Liver Transpl 2015; 21: 881888. 86. Umgelter A, Lange K, Kornberg A, et al. Orthotopic liver transplantation in critically ill cirrhotic patients with multi-organ failure: a  single-center experience. Transplant Proc 2011; 43: 3762-3768. 87. Moreau R. Acute-on-chronic liver failure: a new syndrome in cirrhosis. Clin Mol Hepatol 2016; 22: 1-6. 88. Putignano A, Gustot T. New concepts in acute-on-chronic liver failure: implications for liver transplantation. Liver Transpl 2017; 23: 234-243.

Clinical and Experimental Hepatology Suppl 1/2017

s15

Programme and abstracts*

*The abstracts are printed in the form sent by authors, accepted by the Scientific Programme Committee.

Introductory word The idea of Central European Hepatological Collaboration (CEHC) was developed during informal discussions between hepatologists from Czech Republic, Hungary, Poland and Slovakia in early 2015. The aim of CEHC is to find out the way of possible collaboration between physicians and medical scientists involved in educational and scientific activities related to liver diseases in the Central Europe. We already started research program on long term follow-up of HCV treatment with interferon-free regimens and we hope to begin shortly programs on the HCV screening and hepatic fibrosis. CEHC is also open for collaboration with hepatologists from other countries of the region. The first Meeting of the Initiative Group for Central European Hepatological Collaboration (CEHC) was very exclusive and attended by just eighteen representatives of hepatology associations/sections from the region and took place on 19-21 November 2015 in Warsaw. Further meetings were accompanied CELD Conference in Budapest (23-25 June 2016) and Academy of Contemporary Hepatology in Cracow (6-8 October 2016). The 9th Symposium on Portal Hypertension in Banská Štiavnica (16-17 June 2017) will be an excellent opportunity to meet again, present already available data and discuss further plans of hepatological collaboration in central Europe. Prof. dr hab. med. Robert Flisiak

Clinical and Experimental Hepatology Suppl 1/2017

s19

Dear colleagues, we would like to invite you to the 9th Symposium on Portal Hypertension (SPH-9) with the main topic 'The liver and infections'. The relationship between infections and advanced chronic liver disease (ACLD) has been the hot topic, since infections are the cause or complication of up to 20-30% of ACLDs. Moreover, ACLD-infections connection has exceeded the limits of simple association, and is considered causal. This can be seen in the names of known syndromes (i-ACLF, i-AKI, CAIDS, etc.). To highlight its severity, the cirrhosis-associated immune dysfunction (CAID) was likened to AIDS by the name cirrhosis-associated immune deficit syndrome – CAIDS. Fields of hepatitis B and C treatments are the preeminent examples of the potential of the translational medicine; hepatitis E virus stepped over its percieved geographical borders and should receive its standard role in the differential diagnostic considerations when it comes to acute or chronic hepatitis all around the Europe. This year is very special for SPH because it was given the honour to host summit of the CEHC (Central European Hepatological Collaboration) organization. We hope that the meeting in the Old Castle will be the fertile ground for creating and strenghtening the perpersonal, professional and international relationships with the benefit for our patients. We are looking forward to meeting you in Banská Štiavnica! Lubomir Skladany Svetlana Adamcova-Selčanová

Clinical and Experimental Hepatology Suppl 1/2017

s21

Programme 9 Symposium on Portal Hypertension – The liver and infections th

9.00-12.30 12.30-14.00 14.00-18.00 15.00-15.25

Friday/Piatok, 16 June 2017 CEHC meeting (Kammerhof)/Zasadnutie CEHC CEHC lunch (4 Sochy) Open registration/Registrácia otvorená Sympózium firmy Astellas Clostridium difficile infection in patients with ACLD and after liver transplantation/ Infekcia Clostridium difficile u pacientov s ACLD (advanced chronic liver disease) a po transplantácii pečene

15.30-15.35

Opening of the 9th Symposium on Portal Hypertension (SPH-9)/Otvorenie 9. Sympózia o portálnej hypertenzii

L. Gombošová (UPJŠ, Košice) Predsedníctvo: Š. Hrušovsky – HO pre hepatológiu, I. Schréter – HO pre infektológiu Autori: Ĺ. Skladaný (prezident SHS)

15.35-15.40

Introduction from the President of Slovak Society of Infectology/Slovo prezidenta SSI Pavol Jarčuška (prezident SSI)

15.45-17.30

I. Blok CEHC Predsedníctvo: R. Flisiak, B. Hunyady, P. Jarčuška, J. Šperl

15.45-16.00

Treatment of HCV infection in ACLD and after liver transplantation: The news/Liečba HCV infekcie pri ACLD a po transplantácii pečene: Novinky R. Flisiak (Department of Infectious Diseases and Hepatology, Medical University of Białystok, Poland)

16.00-16.15

Bacterial infections in ACLD: Review/Bakteriálne infekcie pri ACLD: Review B. Hunyady (Pécsi Tudományegyetem, Klinikai Központ, l.sz. Belgyógyászati Klinika, Hungary)

16.15-16.30

Role of serological markers in the diagnosis and prognosis of bacterial infection in cirrhosis/ Úloha sérologických markerov pri diagnostike a prognóze bakteriálnych infekcií pri cirhóze pečene M. Papp (University of Debrecen Clinical Centre, Institute of Internal Medicine, 2nd Department of Medicine)

16.30-16.45

Direct Acting Antiviral (DAA), hepatocellular carcinoma and HBV reactivation: Controversies/ Priamo posobiace antivirotiká (DAA), hepatocelulárny karcinóm a reaktivácia HBV: Kontroverzie

16.45-17.00

Albumin and infections in ACLD: update/Albumín a infekcie pri ACLD: update

17.00-17.10

Infections in ACLD: economic aspects/Infekcie pri cirhóze: ekonomické aspekty

17.10-17.20

Antibiotic stewardship on the Liver Unit/Na výsledkoch založené riadenie antibiotickej liečby v hepatológii

J. Šperl (IKEM, Praha) P. Jarčuška (UPJŠ, Košice) J. Lata (OU, Ostrava) J. Šváč (FDR, Banská Bystrica)

17.20-17.30 17.30-17.45 17.45-19.00

Discussion/Diskusia Coffee break II. Blok

17.45-18.00

Microbiome in ACLD and infections/Mikrobióm ACLD

18.00-18.10

Rare fungal infections in patients with ACLD/CAID and after liver transplantation/ Zriedkavé mykózy u pacientov s CAID a po tronsplantácii pečene

18.10-18.20

Liver abscesses/Abscesy pečene

18.20-18.30

Echinococcosis: the infectologist‘s view/Echinokokóza z pohĺadu infektológa

18.30-18.40

Echinococcosis: the surgeon‘s view/Echinokokóza z pohĺadu chirurga

18.40-18.50

Echinococcosis: the invasive radiologist‘s view/Echinokokóza z pohĺadu invazívneho rádiológa

Predsedníctvo: S. Okapec, D. Pindák, P. Jarčuška L. Bajer (IKEM, Praha)

P. Jarčuška (UPJŠ, Košice) M. Brunčák (FDR, Banská Bystrica) Z. Paraličová (UPJŠ, Košice) M. Oliverius (IKEM, Praha) S. Okapec (FDR, Banská Bystrica)

18.50-19.00 19.00

s22

Discussion/Diskusia Welcome reception/Uvítací večer

Clinical and Experimental Hepatology Suppl 1/2017

Saturday/Sobota, 17 June 2017 III. Blok

9.00-11.15

Predsedníctvo: S. Fraňková, P. Kristián, M. Janičko

9.00-9.45

HIV, HBV, HCV: Slovak permutations/HIV, HBV, HCV: SK Permutácie

9.45-10.00

HBV infection, ACLD and liver transplantation/HBV infekcia, ACLD a transplantácia pečene

10.00-10.15

Secondary hepatotropic viruses and ACLD/Sekundárne hepatotropné vírusy a ACLD

10.15-10.30

Non-hepatotropic virus infections in ACLD/Nehepatotropné vírusové infekcie pri ACLD

10.30-10.45

HAV in ACLD and after liver transplantation/HAV pri ACLD pečene a po transplantacii pečene

10.45-11.15

Hepatitis E: review and situation on Slovakia/Hepatitída E: prehĺad a situácia na Slovensku

P. Jarčuška (UPJŠ, Košice) S. Fraňková (IKEM, Praha) P. Kristián (UPJŠ, Košice) E. Lovrantová (FDR, Banská Bystrica) L. Paraličová, M. Novotný (UPJŠ, Košice) I. Schréter (UPJŠ, Košice), S. Adamcová-Selčanová (FDR, Banská Bystrica)

11.15-11.30 11.30-12.40

Coffee break IV. Blok

11.30-11.45

Diagnosis of sepsis in the era of omics/Diagnostika sepsy v ére “omics“ technologií

11.45-11.55

Infection in the pathogenesis of portal hypertensive bleeding/Infekcie v patogenéze krvácania pri portálnej hypertenzii

Predsedníctvo: P. Trunečka, Š. Hrušovský, E. Lovrantová M. Průcha (Nemocnice Na Homolce, Praha) J. Baláž, M. Brunčák (FDR, Banská Bystrica)

11.55-12.05

The allocation of antibiotic therapy by MIC and penetration: the use of software in ACLD/ Alokácia antibiotickej liečby podĺa MIC a penetrácie pri ACLD za pomoci softvéru

12.05-12.15

Infections after liver transplantation: Review/Infekcie po transplantácii pečene: Review

12.15-12.25

The role of histopathologist in the diagnosis of patients infections after liver transplant patients/ Úloha histopatológa pri diagnostike infekcií u pacientov po tronsplantácii pečene

12.25-12.40

Vaccination in patients with ACLD, before and after liver transplantation/ Vakcinácia u pacientov s ACLD, pred a po transplantácii pečene

A. Purgelová (FDR, Banská Bystrica) P. Trunečka (IKEM, Praha)

E. Honsová (IKEM, Praha)

L. Krištůfková (FVZ – SZU, Bratislava)

12.40-12.55 12.55-13.50

Coffee break V. Blok Predsedníctvo: J. Martínek, S. Dražilová, T. Koller

12.55-13.05

Bacterial infections in ACLD: cohort analysis/Bakteriálne infekcie pri ACLD: Analýza súboru

13.05-13.20

Fungal infections after liver transplantation: cohort analysis/Mykotické infekcie po transplantácii pečene: analýza súboru

13.20-13.30

Infections in primary sclerosing cholangitis (PSC): Review/Primárna sklerotizujúca cholangitída (PSC) a cholangitídy: Review

13.30-13.40

Antibiotic‘s prevention before endoscopic interventions/Prevencia antibiotikami pred endoskopickými zákrokmi

13.40-13.50

CAID(S): ACLD as a severe immune deficit/CAID(S): ACLD ako závažný imunodeficit

13.50-14.00

Concluding remarks/Ukoncenie SPH-9 – záverečné slovo2

14.00-15.00

Lunch at the castie courtyard/Obed no nádvorí zámku

N. Bystrianska (FDR, B. Bystrica) S. Adamcová-Selčanová (FDR, B. Bystrica) J. Martínek (IKEM, Praha) S. Dražilová (NsP, Poprad) K. Kropáčeková, L. Hochmuth (FDR, B. Bystrica) P. Jarčuška, Ĺ. Skladaný

1 Since cirrhosis is an anatomical diagnosis requiring a liver biopsy, the term 'compensated advanced chrnic liver disease' (cACLO) has been suggested rather than cirrhosis (Liver International 2017; 37 (Suppl 1): 104-115) 2 Vyhlásenie troch najlepšich pôvodných prác lekarov do 35 rokov

Clinical and Experimental Hepatology Suppl 1/2017

s23

Oral section Treatment of HCV infection in patients with liver cirrhosis and after liver transplantation Robert Flisiak Department of Infectious Diseases and Hepatology, Medical University of Bialystok, Poland Development of pangenotypic, highly effective and safe IFN-free medications based on combinations of 2-3 direct acting antivirals changed the landscape of HCV treatment. Their combinations provided sustained virologic response (SVR) rates >90% irrespective of liver fibrosis, disease advancement and treatment history. Additionally, these new therapeutic options are safe and allow to shorten treatment to 8-12 weeks in majority patients. IFN-free regimens provided opportunity to cure patients which were previously not allowed for IFN based therapeutic options, such as cirrhotics and liver transplant recipients. According to EASL recommendations (2016) there is no difference between management of patients with compensated liver cirrhosis (Child-Pugh class A) and patients without cirrhosis. Patients with decompensated cirrhosis (Child-Pugh B or C) and MELD < 18-20 can be treated prior to liver transplantation as soon as possible, but protease inhibitors should be avoided in patients classified as Child-Pugh B or C. Patients with MELD ≥ 18-20 should be transplanted first, without antiviral treatment and followed for possible HCV recurrence. All patients with post-transplant recurrence should be considered for therapy, which should be initiated as soon as possible but optimal after 3 months post-transplant. According to the most recent (2017) recommendations of the Polish Group of Experts for HCV, patients with Child-Pugh class C should be primarily recognized as eligible for liver transplantation. Howevre in patients with the MELD ≤ 20 antiviral therapy can be considered before the liver transplantation to suppress viral load to undetectable levels at least a  month prior transplantation to protect transplanted liver from the infection. In patients with MELD > 20 antiviral therapy should be preceded by the liver transplantation, which also applies to patients with too short expected waiting period to ensure effective suppression of HCV. Analysis of available real world experience (RWE) data collected from 13858 patients infected with genotype 1 treated with LDV/SOF ± RBV demonstrated SVR rate of 94% in the whole population and 92% in cirrhotics. OPr ± D ± RBV treatment efficacy analysed in 4260 s24

patients showed SVR rate of 97% irrespective of cirrhosis status. RWE data for other regimens in cirrhotics such as SOF + SMV demonstrated SVR < 90%. Efficacy of LDV/ SOF ± RBV and OPr ± D ± RBV in liver transplant recipients exceeded 90% in several available RWE studies.

Real world experience with interferon-based and interferon-free antiviral therapies against hepatitis C virus in Hungary Béla Hunyady, Michael Makara Somogy County Kaposi Mór Teaching Hospital, Kaposvár, Hungary and University of Pécs, Pécs, Hungary Saint Laslo and Sint Istvan Hospital, Budapest, Hungary Between 2002 and 2015, approximately half of eligible hepatitis C (HCV) infected patients could be effectively treated with pegylated interferon (PegIFN) + ribavirin (RBV) dual therapy to reach a  sustained viral response (SVR). The efficacy has been increased by adding direct acting antiviral drugs (DAA-s: boceprevir, telaprevir or simeprevir) to the PegIFV + RBV regimen in 2013 and 2014. However, IFN-related side effects as well as IFN contraindications have limited the efficacy and/or the applicability of these regimens. Due to financial constraint, the PegIFN + RBV combination is still in use in Hungary as mandatory first line therapy for patients with no contraindication. Data of 774 treatment-naive PegIFN + RBV treated patients starting treatment between 01 August 2013 to 31 July 2015 have been retrospectively collected from the Hungarian Hepatitis Registry. Since genotyping (GT) is not mandatory for IFN-based therapy in Hungary, GT distribution is unknown (however, appr. 90% GT1b based on epidemiology data). Fibrosis staging (FS, also not mandatory) is available for 63% of patients: liver stiffness (LS) < 9.6 kPa in 36%, ≥ 9.6 kPa in 27%, unknown in 37%. An unsatisfactory 35% total SVR rate is predicted (42% – 27% – 37%, according to FS/LS categories mentioned) – although patients with more advanced cirrhosis (decompensated or LS > 20 kPa), or with other negative predictors (HIV co-infection, post liver/organ transplant, relevant baseline cytopenia, specific other co-morbidities) have not been treated with this regimen in Hungary during this period of time. Since 2015, HCV infection (especially genotype 1b, most frequent in Hungary) can be cured with combination of different DAAs (± RBV), without use of IFN, providing an SVR rate of > 95% in almost all patient populations in need. Since May 2015, two of these regimens (ABT3D ± Clinical and Experimental Hepatology Suppl 1/2017

RBV = ritonavir boosted paritaprevir/ombitasvir ± dasabuvir ± RBV or LED/SOF ± RBV = ledipasvir/sofosbuvir ± RBV) have been available in Hungary, too, for a proportion of patients (based on a severity/urgency-driven priority system). With unfortunately > 1500 diagnosed and eligible patients still on a waiting list for these therapies, 912 patients (GT1b = 89.7%; FS < 4: 18%, FS = 4: 60%, FS not known: 12%; ABT3D ± RBV, n: 664, LED/SOF ± RBV, n: 248) have been treated by December 2016 in Hungary with an overall SVR rate of 97% (ABT3D ± RBV: 98.5%, LED/SOF ± RBV: 93.1%). Patients treated with LED/SOF ± RBV had higher average LS (32.9 kPa vs. 25.1 kPa), and included 114 patients (46%) with Child-Pugh B or C stage cirrhosis. In conclusion, IFN-based therapies are significantly underperforming (SVR < 40% in real world experience) and raise safety and/or adherence issues in a  large proportion of patients compared to IFN-free therapies (SVR > 95%). For this reason, IFN-based therapies should not be used for the treatment on any patient – regardless of potential (probably nor existing) financial reasons.

Role of serologic markers in the diagnosis and prognosis of bacterial infection in cirrhosis Maria Papp Department of Internal Medicine, Division of Gastroenterology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary Infectious episodes represent particularly important causes of progression of liver failure and the development of liver-related complications. In cirrhosis, accurate prediction, early recognition and prognostication of bacterial infections are equally essential to surmount complications, delay progression and diminish mortality, however these are challenging in the clinical practice. Serologic markers related to these processes can assist clinicians in decision-making when establish treatment and care strategy for the patients suffering with end-stage liver disease. Pathological bacterial translocation (BT) is increamentally increasing with diseases severity in cirrhosis and has an important role in the development of systemic infections. Inflammatory state sustained by BT is sufficient alone to elevate inflammatory markers to a significant level, even in the absence of without presence of overt infection. Accordingly, of the acute phase proteins (APPs) namely low-grade increases in the level of C-reactive protein (CRP) and lipopolysaccharide binding protein (LBP) were established as risk factors for developing of systemic bacterial infections. Recently, IgA isotype of various serological antibodies directed against gut innate immune proteins or Clinical and Experimental Hepatology Suppl 1/2017

intestinal microorganisms have also been reported as reliable markers of infectious risk in this patient population. Presence of target specific IgA antibodies could be a clue of an excessive mucosal immune response due to extended microbial challenge or dysregulation thereof. In the clinical practice, conventional biomarkers such as C-reactive protein (CRP) and procalcitonin (PCT), are used most frequently in the diagnosis of bacterial infection. However, several diseases specific pitfalls attenuate the diagnostic accuracy of these APPs in cirrhosis. Firstly, if the main source of the APP is the liver, synthesis of protein can be affected by liver failure and its severity. Secondly, depending on molecular weight renal failure and also renal replacement therapy can be confounding factors. Acute kidney injury is frequent in patients with cirrhosis, especially in bacterial infections. Novel markers, such as presepsin or mid-regional pro-adrenomedullin (MR-proADM) have also some drawbacks. Excessive elevation in the level of certain APPs, particularly PCT or anti-inflammatory response related molecules (soluble[s] CD163, soluble urokinase-type plasminogen activator receptor [suPAR], monocyte HLA-DR loss or sTNF-R) are associated with higher risk of short-term mortality during bacterial infections and supposedly represent the deleterious effect of the exaggerated inflammatory processes. Conclusions: Novel biomarkers being devoid of the limitations of classic APPs are highly needed to optimize the rule in and rule out processes necessary for the diagnosis and also for the severity assessment of bacterial infections. Furthermore they may help the identification of patients at high risk for developing systemic infections and those that mostly have advantage from prophylactic measures.

Direct acting antivirals (DAA), hepatocellular carcinoma and HBV reactivation: Controversies Jan Sperl Department of Hepatogastroenterology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic Direct acting antivirals (DAA) combinations have an excellent efficacy and tolerability in contrast to the interferon-based regimens in the treatment of chronic hepatitis C (HCV). However, DAAs have no immunomodulatory effect and the elimination of HCV provides a shift in immunological control in the liver. A decrease in interferon-stimulated genes expression and NK-cells function normalisation in the liver was described by Serti et al. s25

Furthermore, Vilani et al. described an increase of vascular endothelial growth factor serum levels accompanied by the decrease in IL-10 and TNF-α levels. The changes in immunological parameters represent the molecular basis for other viruses resurgence and uncontrolled growth of HCC. In HCV/HBV coinfection, HBV DNA level is often low or undetectable and HCV is usually the main driver of chronic hepatitis activity. HBV may reactivate after HCV eradication. HBV reactivation was described across various DAA regimens, i.e. simeprevir + sofosbuvir (± ribavirin) or ledipasvir/sofosbuvir. 29 cases of HBV reactivation in time relation to DAA initiation were recorded in the FDA database from 11/2013 to 10/2016. Reactivation typically occurred within 4-8 weeks of HCV therapy initiation, and most patients were from Japan (66%) and the rest form the US or other countries (17% each). In these 29 cases, 2 patients died, 1 got liver transplantation, 6 were hospitalized, and 10 discontinued DAAs. Consistently with the above-described, the latest EASL guidance (2016) recommended that these patients should be tested for HBsAg, anti-HBc and anti-HBs. Concomitant HBV therapy is indicated if HBsAg is present or if HBV DNA is detectable in HBsAg-negative, anti-HBc positive patients. HCC occurs in 1-7% patients with cirrhosis per year. The risk correlates with fibrosis stage. SVR is associated with a  reduced mortality and risk of HCC. Conti et al. followed-up 344 individuals with liver cirrhosis for 12-24 weeks after completion of DAA therapy, 59/344 patients had undergone a curative treatment of HCC before therapy. HCC incidence within 24-week period after DAA treatment was 7.6%, in those without HCC 3.2%. In the group with previous HCC, the recurrence rate was 29%. In contrast, in a French study evaluating retrospectively 6000 patients, an increase in HCC incidence after DAA treatment was not confirmed. The patients at risk of HCC should be carefully screened for HCC after DAA therapy by the means of ultrasound every six months.

Antimicrobial stewardship program (ASP) on hepatogastrotransplantation unit (HEGITO) Juraj Svac, Lubomir Skladany, Anna Purgelova, Eva Hruba II Internal Deptartment, F.D. Roosevelt Teaching Hospital, Banská Bystrica, Slovakia Introduction: There are two methods of ASP: 1. Prior authorisation of certain restricted antimicrobials (atb)*. 2. Prospective audit and feedback**. The first aproach leads to reduction of restricted atb but increase use of non restricted drugs. Second aproach helps the physician to

s26

choose the most appropriate therapy and with prospective analysis identificate and eventually correct suboptimal management of infectious disease (ID). This method shows lower atb use, lower number of new atb prescriptions with improved physician satisfaction. The change-over from first to second aproach is multi-step process, which requires: 1. Collect data about every infectious event. 2. Identify most frequent ID in specific departements. 3. Prepare guidelines for their management. 4. Analyse collected data and find if there is right drug choice, right dose, culture directed de-escalation and right duration. Aim: To introduce first experiences with transfer proces from the front-end* to the back-end** ASP aproach on HEGITO. Method: Descriptive statistical analysis of the data (age, gender, reason of hospitalisation, diagnosis of the infectious disease, atb, effect of therapy) extracted automatically from atb indication questionnaire (IQ). IQ should be completed in hospital information system for any formaly restricted atb. Any IQ require prior authorisation by the member of hospital ASP committee. First atb dose was allowed. We analyse the IQs since 1.1.2015 to 31.12.2015. We were recording compliance of ID management with guidelines, finished after this time period, and identify the targets of future ASP intervention. Results: We analysed 192 episodes of ID, in 133 pt., 65 man and 68 woman, with average age 53,5y (19.4y-84y). We have recorded 52 (27%) lower respiratory tract infections (RTI), 50 (26%) blood stream infection (BSI), 29 (15%) billiary tract infection (BTI), 26 (13.5%) spontaneous bacterial peritonitis (SBP), 23 (12%) urintary tract infection (UTI). Empirical atb choices were 47 (33%) chinolons, 39 (20%) piperaciln/tazobactam, 35 (18%) meropenem, 22 (11%) vancomicin, 10 (5%) amikacin, 5 (2.5%) cefoperazon/sulbactam, 5 (2.5%) linezolid. Successful empiric therapy was recorded in (72%) ep. Clincal or laboratory rezistance to empric therapy was in 21%. In hospital mortality on empiric treatement was 14%. Most frequent inhospital mortality was in upper UTI 25% and BSI 16%. Most frequent resistance to empiric atb terapy was in lower RTI 36% and in BTI 29%. Compliance with ASP guidelines: ID drug dose deescalation duration RTI 21% 100% 0% NO DATA BTI 59% 100% 0% NO DATA SBP 41% 100% 0% NO DATA UTI 0% 100% 0% NO DATA Conclusions: ASP on HEGITO demostrate: 1. High resitance to empiric atb choice. 2. Low compliance with subsequently developed guidelines. 3. Overuse of chinolons. Targets for ASP intervention are to increase compliance with drug choice and deescalation.

Clinical and Experimental Hepatology Suppl 1/2017

Gut microbiota and ACLD Lukas Bajer, Pavel Drastich IKEM Praha, Videnska 9, Praha, Czech Republic Current scientific evidence supports that the gut microbiota plays a crucial role in human metabolic and immune homeostasis. As the microbial components and metabolites can cross the gut barrier and enter portal venous blood, the cells located in the liver are constantly exposed to gut-originated substances which interact with large specter of pattern recognition receptors (PRRs) and associated signaling molecules. Advancements in next-generation sequencing technologies during last several years enabled a thorough description of microbial composition in large cohorts of patients with both intestinal and/or extraintestinal diseases. Observed microbial alterations included various liver disorders such as alcoholic liver disease, non-alcoholic fatty liver disease and advanced cholestatic disorders. Several studies established that the liver cirrhosis is, among other distinct features, characterized by overrepresentation of oral microbes in lower levels of gastrointestinal tract.Recent study from Institute for Clinical and Experimental Medicine in Prague (IKEM) indicate that also primary sclerosing cholangitis (PSC) and concomitant inflammatory bowel disease (PSC-IBD) are associated with disease-specific dysbiotic features. These included changes in global microbiota composition, disturbance of gut microbial diversity and altered abundance of specific bacterial taxa. Revealing the specific features of liver disease-associated dysbiosis might contribute to establishing suitable biomarkers predicting the clinical course of advanced chronic liver diseases. Furthermore, deeper understanding of host-microbiota interactions regarding specific microbial taxa might set path towards novel therapeutic targets.

Management of hepatic abscess Michal Bruncak HEGITO – Department of Hepatology, Gatroenterology and Liver Transplantation, F.D. Roosevelt University Hospital, Banská Bystrica, Slovakia Hepatic abscess (HA) remains a serious and often difficult to diagnose problem, because of the nonspecific symptoms and laboratory findings. HAs can be divided into three main categories based on the underlying conditions: infectious, malignant, and iatrogenic. Infectious abscesses include those secondary to direct extension from local infection, systemic bacteremia and intra-abdominal infecClinical and Experimental Hepatology Suppl 1/2017

tions that seed the portal system. However, over the years, the etiologies and risks factors for HA have continued to evolve. Pyogenic HA are relatively rare, though untreated are uniformly fatal. A  recent shift in the management of liver abscesses, facilitated by advances in diagnostic and interventional radiology, coupled with improvements in microbiological identification and therapy, have decreased mortality rates to < 5-30%. Depending on its characteristics, HA can be effectively treated by either percutaneous or surgical drainage in combination with antibiotics. Early recognition of HA is important for instituting effective management and achieving good outcomes.

Echinococcosis: the infectologist‘s view Zuzana Paralicova Department of Infectology and Geographical Medicine, Faculty of Medicine Comenius University and University Hospital in Bratislava, Slovak Republic Echinococcosis is a disease caused by the larval stage of tapeworm from genus Echinococcus. Humans become accidental intermediate hosts. There are two types of echinococcosis – cystic (CE), which is caused by E. granulosus, and alveolar (AE) caused by E. multilocularis. Slovakia is an endemic region of both species. The final host of E. granulosus is a dog for E. multilocularis it is a fox. After ingestion of eggs by intermediate host the released larvae migrate to organs, usually to the liver, where they produce cysts. Cysts caused by E. granulosus are marginated, coated with rigid fibrous membrane. They produce daughter cysts inside the cyst and eventually they calcify. However, they may rupture, what can lead to a  severe allergic reaction and hematogenous seeding. Alveolar cysts are of malignant nature. They are made up of multiple separate vesicles and they are not bounded by fibrous membrane. They grow into the surrounding tissues and form new metastatic cysts in distant organs. Portal hypertension is a rare complication of echinococcosis. Portal hypertension caused by echinococci may be an intrahepatic (in AE), pre-hepatic and post-hepatic (more often with CE). Diagnosis of echinococcosis is based on imaging studies and the serological detection of antibodies. Currently, ELISA assays are the most commonly used. The sensitivity and specificity of individual tests vary greatly and not all commercially available tests are type specific. In practice, we have experience with delayed diagnosis of echinococcosis, often after surgery intervention for a “tumor”, without previous serologic testing. Timely and correct diagnosis is essential for the optimal treatment. Better interdisciplinary cooperation is essential.

s27

Echinococcosis: the invasive radiologist‘s view

Hydatid disease is caused by the larval stages of Echinococcus granulosus. Most patients with hydatid disease have no symptoms, unless there is compression of vital organs such as the hepatic veins, portal vein, hepatic artery in the liver, resulting in life threatening complications. There are four treatment strategies for cystic echinococcosis (CE) – surgery, percutaneous methods, medical treatments and watch and wait strategies. Medical treatment with albendazol or mebendazole may cure only 2/3 of patients with CE. More than 30% of patients will reoccur after stopping the treatment. Watch and wait strategy is followed for asymptomatic and small cysts or CE type 4 and type 5 cysts. For the past two decades, invasive surgery was the recommended standard of practice for the treatment of CE. Today, only complicated cysts, such as biliary fistulae, ruptures in the peritoneum, invasion of the pleural cavity or bleeding into the cyst are surgically treated. Radical and conservative surgical techniques have a higher mortality (2-4%) and morbidity (11-23%) rate, with greater recurrence (2-10.4%) of re-infection and a longer rate of hospitalization than in PAIR (puncture, aspiration, injection, re-aspiration) and Örmeci technique. Both the PAIR and Örmeci techniques are safe and effective. However, the Örmeci technique offers a simpler, inexpensive method of treatment, with no mortality, lower morbidity, low recurrence rate. It can be used as the first choice of treatment modality in patients with cysts type CE type 1, CE type 2, CE type 3A and CE type 3B.

therapies, LT represents now the crucial cure for HBVrelated end-stage liver disease or HCC. Decompensated HBV-related cirrhosis is an uncommon condition and the main indication for transplantation in HBV-infected patients will continue to be HCC. In decompensated HBV cirrhosis, the indication for LT is similar to other causes of cirrhosis. In addition, it is essential to know the precise HBV status of the patient and in particular the existence of HBV replication. Whatever the level of HBV DNA, if detectable, antiviral treatment with entecavir o  tenofovir should be started as soon as possible. The antiviral treatment has two objectives: 1) the improvement of liver function; and 2) to decrease the risk of HBV recurrence after LT since viral replication level at the time of LT correlates with the risk of HBV recurrence, positive HBV DNA at the time of LT seems to influence mortality due to HBV recurrence in HBV/HCC patients. Since interferon treatment is contraindicated in advanced cirrhosis, the only choice for these patients is treatment with NUCs. Lamivudine first and adefovir have been widely used to treat HBV in patients awaiting LT. However, tenofovir and entecavir are currently the first-line drugs with a greater antiviral potency and higher barrier to resistance. In case of previous resistance to lamivudine, tenofovir is the drug of choice, showing a good safety profile even in patients with advanced liver disease. Lactic acidosis has been reported in some patients with MELD score > 20, particularly when treated with entecavir. About one third of patients who initiate therapy have improvement in liver function, which in some cases might result in patient delisting. Cases of severe HBV reactivation should be considered specifically: the treatment with NUCs is an emergency. After LT, lifelong prophylaxis of HBV recurrence in the liver graft based on either hepatitis B immune globulin and NUCs or NUCs alone is the gold standard.

HBV infection, ACLD and liver transplantation

Secondary hepatotropic viruses and ACLD

Sona Frankova

Pavol Kristian

Institute for Clinical and Experimental Medicine, Department of Hepatogastroenterology, Prague, Czech Republic

Department of Infectology and Geographical Medicine, Faculty of Medicine Comenius University and University Hospital in Bratislava, Slovak Republic

Stanislav Okapec FNsP FDR, Banská Bystrica, Slovakia

As a consequence of vaccination and other preventive measures, the epidemiology of HBV infection has stabilized in Western countries. The development of effective antiviral therapies over the years have led to improved survival in HBV patients, reduced HCC incidence as well as improved survival after liver transplantation (LT). Although once considered a contraindication to LT, with introduction of nucleos(t)ide analogues (NUCs) in HBV s28

In addition to the nominal hepatitis viruses (A to E), there are other viruses that can also cause liver inflammation including herpes virus family (EBV, CMV, HSV, VZV...), adenoviruses, rubella, morbilli, parotitis, coxackie and echoviruses. Majority of them causes primary infection in childhood or young age. Liver injury caused by these viruses is very common, usually only mild or modClinical and Experimental Hepatology Suppl 1/2017

erate and of short duration. Fulminant course of infection is possible, but extremely rare. They may not present in chronic form and cannot lead to liver cirrhosis. Some of them, especially CMV, may pose a special risk to patients with chronic hepatitis or cirrhosis of another aetiology (e.g., also in immunocompromised patients). CMV infection represents one of the most frequent opportunistic infections following solid-organ transplantation including liver transplantation. Furthermore, an association between CMV positive donor and negative recipient serodiscordance and severe HCV recurrence in patients undergoing liver transplantation for HCV liver disease was described. Other observation suggests that CMV may represent an important factor for HCV response to interferon treatment in CMV/HCV coinfected patients. Results of a  large retrospective study indicate that CMV seroprevalence in patients with HCC is significantly higher than in patients without HCC, is positively correlated with serum IL-6 levels in cirrhotic patients, and is positively associated with the presence of other hepatotropic viruses such as HCV and HBV. Most of these observations are supported only by a few data. Future studies will be needed to define the role of CMV in ACLD.

Nonhepatotropic viral infection in ACLD Eliska Lovrantova Department of Infectious Diseases, FNsP F.D.R., Banská Bystrica, Slovakia ACLD, which is associated with specific symptoms of the immune dysfunction, is the final stage of chronic liver diseases of various etiology. Both suppression and hyperergy, paradoxically make their concurrent appearance in the system resulting in the increased reactivity on the acute inflammation, which occurs either locally, or more distant. The worst consequence seems to be a  disbalance of proinflammatory and anti-inflammatory processes with a decompensation of ACLD and a development of ACLF. Although some of the triggers causing this decompensation are already identified, 20% to 40% remain to be still unknown. Etiopathogenesis of unexplained conditions may include influence and consequences of certain infections, also known as non-hepatotropic infections. In case of viral infections, we experience the so-called parainfectious hepatopathy or the exacerbation of the chronical, potentially hepatotropic infection occurring during a viral disease. It seems that the main role is played by the Clinical and Experimental Hepatology Suppl 1/2017

immunity system affected by an expansion of T-lymphocytes activated by an extrahepatic viral infection. A good example might be a so-called “collateral damage”, the model of an accompanying parallel liver damage caused by the influenza virus. The related clinical picture indicates several conditions – from a  mild, nonspecific reactive hepatitis, through focal inflammatory processes and fulminant, seronegative hepatitides, to rare fatal form of hepatitis. At the same time, the influenza virus itself has not been identified in the liver. Evidence proves that the influenza virus may cause an exacerbation of the chronical liver disease and can further trigger a rejection of the transplanted liver.The overview of several other viruses that may cause a parallel liver damage is included in the presentation. A  bacterial superinfection of respiratory viral infection is also a complication, which is far from being insignificant. This fact is not only another reason for an early diagnosis and treatment but also justifies further prevention (i.e. vaccination), which respects concurrent viral diseases of patients with ACLD. An early intervention may reduce morbidity and mortality of affected patients and, on the other hand, may positively influence savings in the public health care sector.

Acute hepatitis A in patients with chronic liver disease Martin Novotny, Zuzana Paralicova, Pavol Kristian Department of Infectology and Geographical Medicine, Faculty of Medicine Comenius University and University Hospital in Bratislava, Slovak Republic Acute hepatitis A  (HAV) has mainly an uncomplicated clinical course. In conditions of acute on chronic liver disease reports have indicated contradictory results. Superinfection with hepatitis A virus is generally held to cause acute hepatic failure and higher fatality rates in patients with underlying chronic liver disease, specifically chronic hepatitis B virus (HBV) infection, alcohol abusers but rarely with chronic hepatitis C virus (HCV) infection. There is also possible to find information about reduction or complete eradication of HCV replication in conditions of HAV superinfection in chronic HCV infection. We evaluated the clinical and virological characteristics of hepatitis A virus infection in conditions of chronic liver disease in patients hospitalized at the Department of Infectology and Travel Medicine in Košice. Fulminant HAV is an uncommon disease but according to evidence and our experience is there suspicion for higher incidence in condition of chronic liver disease.

s29

Hepatitis E: overwiew and situation in Slovakia Ivan Schreter, Svetlana Adamcova-Selcanova Department of Infectology and Geographical Medicine, Faculty of Medicine Comenius University and University Hospital in Bratislava, Slovakia HEGITO – Department of Hepatology, Gastroenterology and Transplantology, 2nd Internal Clinic of SZU, FNsP F.D. Roosevelta, Banská Bystrica, Slovakia Hepatitis E virus (HEV) is the most commom cause of acute hepatitis in the developing countries with reduced sanitary conditions and occurs in both sporadic and epidemic forms. High attack rate is typical in young adults. Acute infections are very severe among pregnant women. In Europe for long time it was considered as imported, acute, self limited disease. From the year 2008 in Slovak republic (SR) 98 cases of acute hepatitis E were recognized. All cases except 14 had no traveler history. In our pilot study we found 43.5% (76 from 175) anti-HEV prevalence in general adult population. First evidence about infections among patients with chronic liver diseases are now also available from SR. From group of 589 patients with elevated aminotransferases 65 was positive for serologic markers of hepatitis E. Formerly it had been assumed that HEV is only the cause of acute hepatitis. Over the last few years HEV infection has been recognized as chronic hepatitis in immunocompromised persons (solid organ transplantant recipients, HIV-infected patients, patients with hematologic malignancies).

Diagnostics of sepsis in the era of “omics” technologies Miroslav Prucha Department of Clinical Biochemistry Hematology and Immunology, Hospital Na Homolce, Prague, Czech Republic Option in understanding sepsis pathogenesis and thus in sepsis therapy is linked to the technologies enabling evaluation of gene expression and its regulatory mechanisms in addition to search for new proteins and metabolites produced in septic patients. Genomics, epigenetics, transcriptomics and metabolomics all represent such scientific quest. Numerous studies have attempted to associate genetic markers of genomic variation (polymorphisms) with incidence or outcome of infectious disease and its sequelae in critically ill patients. TNF gene polymorphisms showed s30

association with an increased incidence as well as adverse outcomes in patients with severe sepsis and septic shock (Stuber et al. 1996). Specific allelic variants of the TNF locus are associated with increased susceptibility and adverse outcome of sepsis. Candidate gene studies have been extended towards pro- and anti-inflammatory cytokines like the IL-1 gene family, IL-6 and IL-10 (Fang et al. 1999). Genomic variants of candidate genes involved in pathogen recognition and signal transduction of inflammatory pathways like CD14 and TLRs may also contribute to incidence, severity and mortality of infectious complications in the critically ill (Hubacek et al. 2001; Mansur et al. 2015). Latest findings from a European wide multicentre trial identified variants in the FER gene which are associated with a reduced risk of death from sepsis due to pneumonia (Rautanen et al. 2015). Genes activity varies due to different epigenetics mechanisms. The main principle is gene expression variability without the change in DNA sequence (Phillips 2008). The example of this phenomenon is the bacteria-host interaction. Bacterial-induced epigenetic deregulations may affect host cell function either to promote host defense or to allow pathogen persistence. Thus, pathogenic bacteria can be considered as potential epimutagens able to reshape the epigenome (Bierne et al. 2012). Transcriptomics evaluates messenger RNA levels for genes in specific cells or tissue. Transcriptomics is used in diagnosis of sepsis for discrimination between infectious and non-infectious inflammation (Prucha et al. 2004; Tang et al. 2010). It can also identify biomarkers for assessment of pathogenetic course and prognosis of the disease (Davenport et al. 2016). The main goal of proteomics is to identify proteins, biomarkers which are produced in septic patients and thus can help in accurate diagnosis of sepsis (Paugam-Burtz et al. 2010), or to detect proteins and to describe their function on the molecular level in sepsis (Buhimschi et al. 2011). The term metabolom includes intra and extracellular low molecule substances which are the product of metabolic pathways needed for cell growth and its function. Metabolomics is complete analysis of metabolom in given physiological or pathological state of the cell, tissue and organism and it provides new insight on cellular fiction (Goodacre et al. 2004). It can be used in septic patients for diagnosis, disease prognosis and for the patient’s risk stratification (Ferrario et al. 2016; Kauppi et al. 2016).

Clinical and Experimental Hepatology Suppl 1/2017

Infections in the pathogenesis of bleeding in portal hypertension Jozef Balaz, Michal Bruncak HEGITO – Department of Hepatology, Gastroenterology and Transplantology, 2nd Internal Clinic of SZU, FNsP F.D. Roosevelta, Banská Bystrica, Slovakia Infectious complications significantly increase mortality in patients with end-stage liver disease and are considered a major medical challenge. Higher incidence of systemic infections, deficiencies in immune system caused by liver dysfunction and bacterial translocation from digestive tract represent the main identified causes of infections in cirrhosis. The pathophysiological mechanisms initiated by infection lead to reduced intrahepatic vasodilatation and increased intrahepatic vasoconstriction, and hence result in increased risk of acute variceal bleeding. Identification of mechanisms and signal molecules involved in the process offers possibilities of new diagnostic and therapeutic approaches to portal hypertension.

Infections in liver transplant recipients: A review Pavel Trunecka IKEM, Prague, Czech Republic Infections are very important cause of morbidity and mortality of the host at all time periods after liver transplantation, being the most common complication with incidence rate 20-80%. They could be divided into 4 groups: 1) infections transmitted by donor organ, 2) infections associated with the surgery (= infections of early post-transplant period), 3) late opportunistic infections of immunocompromised host, 4) reinfections of HBV or HCV positive recipient. Immunosuppressive therapy as well as advanced liver failure contribute to susceptibility of the recipient, most importantly to infections of the 2 group. 1) Most common infections transmitted by graft are bacterial and yeast infections. Multidrug resistant pathogens could be transmitted, esp. if donor was longterm hospitalized and ventilated, and could cause severe sepsis in recipient. Transmission of hepatitis B, C, and E viruses is limited by donor testing, but still possible. Transmission of parasites (Strongyloides, Tripanosoma cruzi) was described. Graft from donor tested positive for Treponema pallidum may be transplanted but recipient must be treated with Penicillin. Clinical and Experimental Hepatology Suppl 1/2017

2) This type of infectious complications is caused mostly by Gram- bacteria (originated mostly from colonized host), or by nosocomial infection (often also Gram+). Multidrug resistant pathogens (MRSA, VRE, ESBL+) could cause serious problems. Most programs employ short-term ATB prophylaxis according to local practice, all cases of high risk recipients (acute liver failure, re-transplantation, high blood loss, patients with kidney failure, and reoperations should be covered by antifungal prophylaxis. All patients in early post operation period are protected by prophylactic treatment for Pneumocystis, occasionally for Legionela pneumophilla. CMV, Tuberculosis, and Toxoplasma require specific protocols of prophylaxis. 3) It is difficult to draw line between infections of early and late post-transplantation period, agents of group 2 could cause complications any time after transplant. The most typical opportunistic infections are caused by Listeria, Nocardia, herpetic viruses (CMV, EBV, HVV6, Varicella zoster virus). Especially serious are infections caused by Cryptococcus, Mucormycosis, and Aspergillus. 4) Hepatitis C recurrence could be challenging even in era of DAA if rare viral resistance is present. Post-transplant prophylaxis for HBV must be continuous. In transplant candidates and recipients, proper vaccination is required. Live vaccines are contraindicated in most cases. Infections after transplantation represents still major challenge and require fast and effective treatment measures in line with institutional guidelines for antimicrobial treatment and prophylaxis, that must be timely updated according to results of local microbial surveillance.

The role of histopathologist in the diagnosis of infections in patients after liver transplantation Eva Honsova IKEM Praha, Videnska 9, Prague, Czech Republic Despite advances in liver transplantation, morbidity and mortality due to infectious complications represent still serious problems. From the clinical point of view, it is useful to divide the post-transplant course into three time periods related to the risks of infection by specific pathogens: the early period post-transplant (first month), an intermediate period (1 to 6 months), and more than 6 months. In the first month post-transplant, there are two major causes of infection in liver transplant recipient: infection derived from either the donor or recipient and infectious complications of the transplant surgery and hospitalizas31

tion. Donor-derived infections has increased with the emergence of antimicrobial resistance. Recurrent viral hepatitis may reemerge early after transplantation. Infectious complications related to surgery represent the common postoperative complications. Bacterial infections predominate; they usually have a nosocomial source. In the period 1 to 6 months post-transplant, the patients are most at risk for the development of opportunistic infections, although residual problems from the perioperative period can persist. Viral pathogens, particularly the herpes group viruses but also de novo and/or recurrent hepatitis B and hepatitis C can occur. Two clinically and morphologically different types of B and C hepatitis are known, one with conventional morphology. The second, unique to the allograft termed fibrosing cholestatic hepatitis, with extensive hepatocellular damage and progressive development of liver failure. Recently, hepatitis E has been recognized more frequently as a cause of graft damage. Incidence and prevalence of hepatitis E post-transplant remains unclear but is certainly greater than historical estimates. More than 6 to 12 months after transplantation, most patients are receiving stable and reduced levels of immunosuppression. These patients are subject to community-acquired pneumonias or severe illness from community-acquired infections. Recipients who have less than adequate graft function tend to require higher than usual immunosuppressive therapy. As a  result, they represent a subgroup of transplant patients at highest risk for opportunistic infections typically encountered during the period of 1 to 6 months after transplant. They are also at risk for the late effects of viral infections manifest as malignancy: post-transplant lymphoproliferative disorder or squamous cell cancers of the skin or anogenital region.

Incidence and consequences of bacterial infections in patients with decompensated chronic liver disease – cirrhosis Natalia Bystrianska, Svetlana Adamcova-Selcanova, Jana Vnencakova, Tomas Koller, Daniela Jancekova, Jana Badinkova, Pavol Molcan, Juraj Svac, Lubomir Skladany HEGITO – Department of Hepatology, Gastroenterology and Transplantology, 2nd Internal Clinic of SZU, FNsP F.D. Roosevelta, Banská Bystrica, Slovakia 5th Internal Clinic, FNsP Ružinov, LF UK, Bratislava, Slovakia Introduction: Over the last 10 years, the overall mortality due to the consequences of liver cirrhosis (suggested to be called advanced chronic liver disease, ACLD) s32

has increased significantly and this is a continuous trend; in Slovakia it ranks 5th among all causes of mortality and Slovakia ranks 4th in Europe in this parameter. The mortality is greatly (estimated at 1/3) affected by infections; the infections are the main reason for decompensation of ACLD (dACLD) on one hand and on the other they are a  complication of dACLD, as well as the cause of death. The significance of infections in the outlook and pathogenesis is so great that “i” (infection) is added to the names of new syndromes (iACLF) or new definition of existing syndromes (iAKI). Aim: Register and observational study HEGITO 7 to enable the evaluation of the association between infection and dACLD in the population of hospitalized patients at HEGITO. Methods: Retrospective analysis of data in electronic database of NIS CareCenter Copyright 2000, CGM version 3.19.1 in patients with ACLD hospitalized at HEGITO BB between July 2014 and December 2015. Inclusion criteria: 1. ACLD; 2. Hospitalization at HEGITO; 3. Informed consent. Exclusion criteria: 1. Malignancy; 2. ACLD following orthotopic liver transplant (OLTx); 3. FMODA (Frequent low-volume ascites drainage through implanted peritoneal catheter); 4. Insufficient data Observed variables: gender, age, etiology of ACLD, ChildPugh score (CPS), MELD (Model for End Stage Liver disease), type of infection in ALCD, mortality. Results: The study population included a total of 235 observed patients (pts); inclusion criteria were satisfied by 217 pts (92.3%); 67 pts (30.9%) had a  confirmed infection complication, in the remaining 150 pts (69.1%) no infection was diagnosed. In the population of patients with infection, there were 42 males (62.7%); with mean age of 54.3 years; 25 females (37.3%) with average age of 57.3 years. The observed population of patients with infection was dominated by etiological factor ALD (alcohol liver disease) 42 pts (62.7%); Child-Pugh score (CPS) was 10.34 ± 1.77 points; MELD score 18.46 ± 8.44 points. In the observed population of patients with infection, 25.3% pts had chronic decompensation (CD), 38.8% pts had acute decompensation (AD) and 73.3% pts had acute-onchronic liver failure (ACLF). The occurrence of infections was as follows: with CD – prevailing infections of urinary system (35.7%), in AD group – spontaneous bacterial peritonitis (SBP) (19.4%) and in the ACLF group – sepsis (26.7%). In the population of patients with infection, the mortality during hospitalization was 14.9% (10 pts); while in the population of patients without infection, the mortality was 1.3% (2 pts) (p = 0.001). Conclusions: Microbial infections represent a major problem in patients with ACLD. In the analyzed population of ACLD patients, we confirmed 30% prevalence of infections .The analysis failed to confirm the effect of gender, age and etiology on the development of infection Clinical and Experimental Hepatology Suppl 1/2017

complications in the observed population of patients (p = NS), just the opposite – statistically significant association (p = 0.0001) was confirmed with respect to CPS and MELD. The greatest occurrence of infections was observed in the subgroup of patients with ACLE, as confirmed by the available publications where the bacterial infections are considered the most common initiating factor.The analysis also demonstrated a significant association between the infections and mortality, underlining their importance in this respect.

Fungal infection after liver transplantation: cohort analysis Svetlana Adamcova-Selcanova, Anna Purgelova, Lubomir Skladany HEGITO – Department of Hepatology, Gatroenterology and Liver Transplantation, Department (Dpt) Internal Medicine II, Slovak Medical University, F.D. Roosevelt University Hospital, Banská Bystrica, Slovakia Introduction: Solid organ transplant (SOT) recipients have a significantly increased risk of fungal infections (FI). They are caused mainly by Candida spp., to a lesser extent by Aspergillus spp. During the past decade, non-albicans spp., like C. glabrata, emerged as an increasingly important causes of FI. Standard protocol of FI prophylaxis after liver transplantation (LTx) with fluconazole (FLU) has been declared ineffective.Careful examination of risk factors, diligent surveillance for early signs of infection, and prompt intervention are the mainstays of FI prevention. Due to the paucity of clinical trials there are no definitive recommendations on the diagnosis, treatment and prevention of FIs in SOT. Aims: To determine 1) the prevalence of FI; 2) type of FI; 3) efficacy of FLU prophylaxis; 4) sensitivity to other antifungals; 5) lenght of hospital stay (LOS) 6) in hospital mortality(IHM) in LTx patients (pts) Transplant Center (TC) BB. Methods: Retrospective analysis. The search of the Hospital Information System “Care Center®” and the database of the Department of Microbiology for the data of consecutive pts with LTx. Susceptibility testing was performed for FLU, itraconazole (ITR), voriconazole (VOR), other azoles and echinocandins. Study interval: May 2008 – January 2017. Inclusion criterion: LTx in TC BB. Exclusion criterion: LTx in other TC. Recorded variables: age, gender, agents of FI, sensitivity to FLU and other antifungals, MELD, LOS, IHM. Results: During study interval 166 pts were enrolled, 95 men (57%), median age 55 years (17-76). FI was recordClinical and Experimental Hepatology Suppl 1/2017

ed in 116 pts (69%), in 155 cases. Candida spp. was present in 152 cases (98%), Aspergilus spp. in remaining 3 (2%). From all FI, Albicans spp. were identified in 91 (60%), 61 (40%) – non-albicans. The most common spp. was C. albicans – 91 (59%), followed by C. glabrata – 34 (22%), C. krusei – 9(6%), C. kefyr – 5 (3%), C. parapsilosis – 5 (3%), C. tropicalis – 4 (3%), C. lusitaniae – 4 (2%) and Aspergillus – 3 (2%). C. albicans was sensitive to FLU in 85 cases (95%), resistant in 5%; C. glabrata was sensitive to FLU in 24%, VOR – 98%, ITR – 1.85%, echinocandis – 100%; C. krusei was resitant to FLU in 100%, sensitive to VOR – 92.8%, ITR – 35.7%, echinocandins – 100%; C. parapsilosis was sensitive to FLU in 40%, other Candida spp. were sensitive to FLU, VOR and echinocandins in 100%. Median LOS in pts with FI was 34 days (d) (0-192), IHM – 11 (9%) and MELD (Model for End Stage Liver Disease) – 17.7 (9-33). Median LOS in pts without FI – 26.5 d (0-90), IHM – 2 (4%), MELD – 16.7 (8-43). Conclusions: FI due to yeast are the most common infections after SOT. Candida spp. was the most common FI in pts after LTx at TC BB. The prophylaxis with FLU is effective in Albicans spp.; in non-albicans spp., the sensitivity is suboptimal. In pts with FI the LOS and IHM was higher. The incidence of FI in our cohort was indepentend of MELD score. Adequate prophylactic strategies with echinocandins can decrease the risk of FI.

CAIDS – ACLD as a severe immunodeficiency Kristina Kropacekova, Ludek Hochmuth, Lubomir Skladany, Jana Vnencakova HEGITO – Department of Hepatology, Gastroenterology and Transplantology, 2nd Internal Clinic of SZU, FNsP F.D. Roosevelta, Banská Bystrica, Slovakia Liver as a primary immunological organ contributes the functioning of the immune processes through its “immunosurveillance“ role and through the synthesis of proteins mediating immune reactions. Thus every liver damage leads to immune dysfunction. This recent syndrome known as CAIDS (cirrhosis associated immune dysfunction syndrome) is a major factor in pathogenesis of the infectious complications in ACLD. CAIDS is a  severe dysfunction of both innate and adaptive immunity and a dysregulation in proinflammatory and counterinflammatory reactions. The CAIDS phenotypes vary from mainly proinflammatory state (exposition to DAMPs from damaged hepatocytes and PAMPs from increased bacterial translocation) through immunodefis33

ciency state to the exhaustion of the immune system, immune paralysis and indolent dead. The immunomodulatory therapy possibilities are not well known yet and offers large field for the future study. Our project is observing the possible effect of transfer factor (TF) on the incidence of infections and mortality in patients with ACLF (acute on chronic liver failure). It is a retrospective, case-control (propensity matched) study.

Poster section Liver abscesses – an increasing problem (analysis of Derer’s University Hospital in Bratislava) Tereza Hlavata, Zuzana Duranova, Ivan Vojtech, Lukas Gregus, Jozef Sedlacko, Maria Szantova 3rd Department of Internal Medicine, Faculty of Medicine, Comenius University and University Hospital in Bratislava, Slovak Republic Department of Infectology and Geographical Medicine, Faculty of Medicine Comenius University and University Hospital in Bratislava, Slovak Republic Introduction: The incidence of liver abscesses (LA) has been still rising over the past years. According to the etiology, bacterial (pyogenic), parasitic and mycotic abscesses may occur. Pyogenic liver abscesses are uncommon conditions that present diagnostic and therapeutic challenges to physicians. However, pyogenic abscesses (PA) are more common among patients at internal departments and mycotic are frequent in hemato-oncological patients. Aim: To analyse the occurrence of liver abscesses in inpatients at 3rd Department of Internal Medicine and Department of Infectology and Geographical Medicine over the period of 5 years. Etiology, microbial origin, localization, type, length and outcome of the treatment were assessed. Results: Pyogenic abscess was present in 11 patients. The mean age of patients was 57 years. The ratio of solitary to multiple abscesses was 6 : 5. The right localization (72.7% patients) was more common in comparison to the left (18.2%) or both lobes (9.1 %). In 4 patients (36.4%), there was biliary etiology and in 1 patient secondary infection of HCC (hepatocellular carcinoma) was present. Etiology of 6 patients remained unexplained. The most common microbial agent was Klebsiella pneumoniae. Positive blood or abscess´s cultivation was present in 6 patients (54.5%). Fever, nausea, vomiting and abdominal pain were most common clinical presentations. Inflammatory process was s34

detected by laboratory findings, and imaging methods were helpful in detecting of their localization. All patients were treated with a parenteral combination of at least 2 types of antibiotics in the period of 6-9 weeks. Percutaneous drainage (PD) was made in 2 patients, repeated unsuccessful effort of PD was made in 1 patient, 6 patients did not need PD. 2 patients needed a  surgical approach. In 3 patients death occurred due to sepsis and multiple organ failure. Remaining 8 patients were successfully treated. Clinical symptomatology disappeared by 8 weeks and PA disappeared by 4-6 months from the start of the treatment. Discussion: Diagnosis of LA is not easy. The clinical presentation is not specific. Establishment of the diagnosis, from the onset of first symptoms, may vary from 2 weeks to 6 months. Fluidothorax or positive auscultation findings are common signs. The etiology of abscess (biliary disease, bacteremia, abdominal inflammation) may be present in a clinical picture. Laboratory findings together with clinical findings are crucial for detection of a serious infection and start of an antibiotic treatment. Imaging methods are key to localize of LA. A correlation of clinical and laboratory findings with an adequate sequence of imaging modalities is important in a clinical assessment. Early start of antibiotic treatment with PD may cure the patient. Conclusions: Nowadays, liver abscesses are an increasing problem with multifactorial etiology and difficult diagnosis and treatment. The treatment has become more effective with a  decrease in mortality over the last years. The possible causes of failure of a treatment involve mainly an increase of antibiotic resistance. Patient's management should be done in cooperation with the internist, infectologist, surgeon, and radiologist.

Acute-over-chronic liver failure (ACLF) in 305 HEGITO inpatients D. Jancekova, L. Skladany, S. Adamcova-Selcanova HEGITO – Division of Hepatology, Gastroenterology and Liver Transplantation, Department of Internal Medicine II of Slovak Medical University, F.D. Roosevelt University Hospital, Banská Bystrica, Slovakia Introduction: ACLF – the new syndrome distinct from both chronic (CD), and acute liver failures. According to the landmark CANONIC study, ACLF is defined by baseline advanced chronic liver disease (ACLD), precipitating insult, acute decompensation (decompensating event present < 2-4 weeks), and is characterized by high short-term (28-day) mortality. It is associated with infecClinical and Experimental Hepatology Suppl 1/2017

tions in around 30% of patients (i-ACLF), whereby iACLF has higher in-hospital mortality. Aim: To determine the prevalence and the main characteristics of ACLF in patients admitted with decompensated ACLD, with special accent on iACLF. Material and methods: Retrospective analysis (DJ). Consecutive inpatients. Inclusion criteria: admission to HEGITO with dACLD. Exclusion criteria: malignancy; insufficient data. Recorded variables: Age; gender; etiology of dACLD; type of dACLD: AD (ACLF 0-3 at admission and at day 7), CD; iACLF at admission; in-hospital mortality. Results: During the period 7/2014 – 04/2016, 305 patients with dACLD were enrolled, 12 (4%) excluded, 215 with CD (70%), 78 with AD (26%). Median age was 56 years; female were 35n (45%); etiology with alcoholic liver disease (ALD) 65 (83%), autoimmune syndromes 11 (14%),other 2(3%). Decompensating events: ascites 55%, upper-GI bleeding 29%, hepatic encephalopathy 8%, infections 8%. Insults: acute alcoholic hepatitis 37%, GI bleeding 27%, bacterial infections 24%, unknown 9%, TIPS 3%. ACLF 1-3 was present in 35% (1 – 17%, 2 – 14%, 3 – 7%); iACLF – 24% of AD patients (SBP – 37%, respiratory – 21%, UTI – 15%, others-upt to 100%). Mortality in the AD group was 12%, as compared to 5% in CD group. Mortality in ACLF grades 1-3 was 18%, 50%, and 80%, respectively. Conclusions: Prevalence of AD in patients hospitalized with dACLD was 26%, ACLF 1-3 was present in 35% of them. Mortality according to ACLF grades correlated with the data from literature. Infections played important role either as the insult, or event; small number of patients with iACLF precluded deeper analysis. Prospective collection of data is underway.

Bacterial infections and hepatic encefalopathy Jan Strachan, Natalia Bystrianska, Jana Vnencakova, Svetlana Adamcova-Selcanova, Daniela Jancekova, Lubomir Skladany HEGITO – Departement of Hepatology, Gastroenterology and Liver Transplantation, F.D. Roosevelt University Hospital, Banská Bystrica, Slovakia Introduction: Bacterial infections are common complication of patients with liver cirrhosis. Moreover, bacterial infections are frequent precipitating factor of hepatic encefalopathy. Infections are asociated with hepatic encefalopathy in 35% - 47% cases and getting worse prognosis of patient with liver cirrhosis. Clinical and Experimental Hepatology Suppl 1/2017

Aim: Find out incidence of bacterial infections and asociation with hepatic encefalopathy in patients with ACLD. Material and methods: Retrospective analysis from Hospital information system CareCenter Copyright 2000, CGM verzia 3.19.1 in patient with ACLD hospitalised on department HEGITO since 7/2014 until 12/2015. Inclusion criteria: 1. ACLD; 2. Hospitalisation on department HEGITO; 3. Informed consent. Exclusion criteria: 1. Malignancy; 2. ACLD after oLTx; 3. FMODA (Frequent small volume paracentesis via CAPD catheter); 4. Lack of information. Monitored variables: gender, age, etiology of ACLD, Child-Pugh score, MELD, hepatic encefalopathy, type of infection. Results: Inclusion criteria fullfiled 235 patiens (pts), 18 pts were excluded, into defenitive analysis were included 217 pts. 67 pts (30.9%) had infectious complication, 150 pts (69.1%) were without infection. In the group pts with infection were 42 men (62.7%); with avarage age 54.3 years, 25 women (37.3%) with avarage age 57.3 years, the most common reason of cirrhosis was Alcohol liver disease (ALD) in 42 pts (62.7%), avarage Child-Pugh score was 10.34 ± 1.77 and avarage MELD 18.46 ± 8.44. In 32 pts with infection (47.76%) was diagnosed overt hepatic encefalopathy. The most common precipitating factor of hepatic encefalopathy were uroinfections (32.4%), SBP (21.6%) and infections of respiratory tract (16.2%). Conclusions: One of the most common precipitating factor of hepatic encefalopathy are infections. In our retrospective analysis we finded out, that 48% pts with liver cirhosis and bacterial infection had also overt hepatic encefalopathy. The most common precipitating factor of hepatic encefalopathy were uroinfections, SBP and infections of respiratory tract.

Role of inflammasomes in liver diseases Maria Szantova, T. Hlavata, Z. Durkovicova, M. Szamosova 3rd Department of Internal Medicine, Faculty of Medicine Comenius University, Bratislava, Slovakia Introduction: A  new term ‘inflammasomes’ has appeared in the literature over the last years. Aim: Clarification of clinical significance of inflammasomes in chronic liver diseases. Material and methods: Analysis of the review of the literature on this topic in the last eight years. Results: Inflammasomes are molecular platforms activated upon cellular infection or stress that trigger the maturation of proinflammatory cytokines such as inters35

leukin-1b to engage innate immune defenses. They are multiprotein complexes responding to pathogen-associated molecular patterns (PAMP) and causing damage to them (DAMP-damage associated molecular patterns) and activate the inflammatory process. Various pathogens (toxins, alcohol, fat, viruses, etc.) may be activators of inflammasomes, most common due to alterations in the gut microbiota or increased intestinal permeability through toll-like receptors (TLR). Inflammasomes are regulators of inflammation and cell death. They are expressed and likely functionally active in the liver in hepatocytes, liver sinusoidal endothelial cells, hepatic stellate cells and macrophages. PAMPs are mostly derived from the gut, due to alterations in gut microbiota composition and/or increased intestinal permeability. DAMPs are mostly derived from damaged hepatocytes and include ATP, uric acid, cholesterol crystals, DNA fragments, and fatty acids. The inflammasome can be activated either directly in hepatic stellate cells, Kupffer cells, and hepatocytes, or indirectly through cell death and increased exposure to PAMPs. Two steps are involved in inflammasome activation. The first step is signalization through TLR in bowel or IL-1 receptor with the following expression of inflammasome patterns. The second step includes releasing of caspase-1 with cytokine activation and secretion. Activation of inflammasomes culminates in caspase-1 activation and IL-1β secretion. IL-1 production has an auto-regulatory loop. The secreted active IL-1β or IL-1α can activate the IL-1 receptor complex and increase the transcription of its own precursor as well as the synthesis of the inflammasome components. This amplification loop suggests that small amounts of IL-1β could have a significant biological effect. In addition to inflammation, inflammasome activation regulates cell death. NLRC4 (NOD-like receptors C4) and NAIP- receptors activate pyroptosis, while NLRP3 (NODlike receptors P3) activation contributes to pyronecrosis. Pyroptosis is a  caspase-1-dependent cell death showing similarities to apoptosis and DNA damage. Unlike apoptosis, pyroptosis does not depend on apoptotic caspases, and it is accompanied by a loss of plasma membrane integrity and lack of chromatin condensation. Pyronecrosis shows similarities to necrosis since it is not caspase-dependent and leads to the breakdown of the plasma membrane without chromatin condensation. There is increasing evidence that gut microbiota, increased gut permeability, and endotoxin contribute to the pathogenesis of both alcoholic (ASH) and non-alcoholic steatohepatitis (NASH). Endotoxin, a  cell wall component of Gram-negative bacteria, is a  major mediator of sepsis-induced liver damage, multiorgan failure, and chronic liver disease. Owing to the portal blood supply

s36

arriving from the intestines, the liver is exposed to high concentrations of nutrients and gut-derived substances including endotoxin. In ALD, serum levels of IL-1β are increased. Potential molecular triggers for inflammasome activation in NASH include DAMPs such as DNA, saturated fatty acids, and PAMPs. In primary hepatocytes, saturated, but not unsaturated fatty acids induce caspase-1 activation and IL-1β release in the presence of endotoxin. Danger signals from fatty acid-treated hepatocytes can induce inflammasome activation in liver mononuclear cells, suggesting a  crosstalk between liver parenchymal cells and immune cells in NASH. This intercellular crosstalk provides potential amplification of inflammasome activation and inflammatory pathways in NASH. Fatty acid-induced inflammasome activation in macrophages is NLRP3-dependent and involved increased mitochondrial ROS production and decreased autophagy due to reduced AMPK activity. Chronic liver inflammation that is amplified by IL-1 leads to fibrosis and cirrhosis. Conclusion: Inflammation is a  common feature of chronic liver diseases (ALD, NAFLD, drug-induced liver injury), diabetes mellitus and atherosclerosis. Inflammasomes and cytokines are the source of sterile inflammation in the liver resulting in fibrogenesis. Inflammasomes and IL-1 participate in all diseases with the autoinflammatory response and metabolic stress and may be related to complications. There is evidence that IL-1β, IL-18, and inflammasomes may also mediate encephalopathy. Caspase-1 and IL-18 levels are higher in patients with acute or acute-on-chronic liver failure compared to controls or patients with stable chronic liver diseases. Quite new therapeutic consequences are expected in the future on the basis of inflammasome inhibition and regulation of programmed cell death (necroptosis). Bibliography Szabo G, Csak T. Inflammasomes in liver diseases. J Hepatol 2012; 57: 642-654. Wree A, Marra F. The Inflammasome in liver disease. J Hepatol 2016; 65: 1055-1056.

Clinical and Experimental Hepatology Suppl 1/2017